1
|
Lee YM, Hsu CL, Chen YH, Ou DL, Hsu C, Tan CT. Genomic and Transcriptomic Landscape of an Oral Squamous Cell Carcinoma Mouse Model for Immunotherapy. Cancer Immunol Res 2023; 11:1553-1567. [PMID: 37669022 PMCID: PMC10618654 DOI: 10.1158/2326-6066.cir-23-0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 06/14/2023] [Accepted: 08/30/2023] [Indexed: 09/06/2023]
Abstract
The immune checkpoint inhibitor (ICI), anti-programmed death-1 (anti-PD-1), has shown moderate efficacy in some patients with head and neck squamous cell carcinoma (HNSCC). Because of this, it is imperative to establish a mouse tumor model to explore mechanisms of antitumor immunity and to develop novel therapeutic options. Here, we examined the 4-nitroquinoline-1-oxide (4NQO)-induced oral squamous cell carcinoma (OSCC) model for genetic aberrations, transcriptomic profiles, and immune cell composition at different pathologic stages. Genomic exome analysis in OSCC-bearing mice showed conservation of critical mutations found in human HNSCC. Transcriptomic data revealed that a key signature comprised of immune-related genes was increased beginning at the moderate dysplasia stages. We first identified that macrophage composition in primary tumors differed across pathologic stages, leading to an oncogenic evolution through a change in the M1/M2 macrophage ratio during tumorigenesis. We treated the 4NQO-induced OSCC-bearing mice with anti-PD-1 and agonistic anti-CD40, which modulated multiple immune responses. The growth of tumor cells was significantly decreased by agonistic anti-CD40 by promoting an increase in the M1/M2 ratio. By examining cross-species genomic conservation in human and mouse tumors, our study demonstrates the molecular mechanisms underlying the development of OSCC and the regulation of contributing immune-related factors, and aims to facilitate the development of suitable ICI-based treatments for patients with HNSCC.
Collapse
Affiliation(s)
- Yi-Mei Lee
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
- Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Hsin Chen
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
- Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Da-Liang Ou
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- YongLin Institute of Health, National Taiwan University, Taipei, Taiwan
| | - Chiun Hsu
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Ting Tan
- Department of Otolaryngology, National Taiwan University Hospital, Taipei, Taiwan
- Stem Cell Core Laboratory, Center of Genomic Medicine, National Taiwan University, Taipei, Taiwan
- Department of Otolaryngology, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Otolaryngology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| |
Collapse
|
2
|
Raabe O, Birchler T, Rehrauer H, Eppler E. CD40 Agonist Monoclonal Antibody-Mediated Hepatitis in TNF-Receptor 1 Gene Knockout Mice. Biomedicines 2021; 9:biomedicines9080863. [PMID: 34440067 PMCID: PMC8389574 DOI: 10.3390/biomedicines9080863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/06/2021] [Accepted: 07/14/2021] [Indexed: 11/17/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) plays an important role in liver inflammation. CD40-CD40 ligand (CD40-CD40L) is a key receptor–ligand signaling pair involved in the adaptive immune response and pathogenesis of autoimmune diseases. In mice, CD40 activation leads to sickness behavior syndrome (SBS) comprising weight loss, sleep disruption and depression, which can be blocked by administration of the TNF-inhibitor etanercept. In the present study, we assessed the extent of hepatic inflammation in mice devoid of the TNF-receptor 1 (TNFR1)-mediated signaling pathway. The TNFR1-depleted (TNFR1−/−) adult mice and their wild type littermates were given a single intra-peritoneal injection of CD40 agonist monoclonal antibody (mAb) or rat IgG2a isotope control. As described previously, TNFR1−/− mice were protected from SBS upon CD40 mAb treatment. Cd40, tnf and tnfr1 mRNA and Tnf-α peptide were increased in the liver of CD40 mAb-stimulated wild type mice. Serum alanine aminotransferase was elevated in both CD40-activated wild type and TNFR1−/− mice. TNFR1−/− mice showed much less intra-parenchymal infiltrates, hepatocellular necrosis, and perivascular clusters upon CD40 mAb activation than their wild type littermates. A gene expression microarray detected increased activity of metabolic and detoxification pathways and decreased activity of inflammatory pathways. We conclude that immune activation and development of liver inflammation in CD40L interactions depend on TNFR1-mediated signaling pathways and are counteracted by alterations in metabolic pathways.
Collapse
Affiliation(s)
- Oksana Raabe
- Department of Biomedicine, University of Basel, 4056 Basel, Switzerland;
| | - Thomas Birchler
- Institute of Experimental Immunology, University of Zurich, 8057 Zürich, Switzerland;
| | - Hubert Rehrauer
- Functional Genomics Center Zurich (FGCZ), ETH Zürich, University of Zurich, 8057 Zürich, Switzerland;
| | - Elisabeth Eppler
- Institute of Anatomy, University of Zürich, 8057 Zürich, Switzerland
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
- Correspondence: ; Tel.: +41-31-684-84-53
| |
Collapse
|
3
|
Line-selective macrophage activation with an anti-CD40 antibody drives a hemophagocytic syndrome in mice. Blood Adv 2021; 4:2751-2761. [PMID: 32559293 DOI: 10.1182/bloodadvances.2020001624] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/18/2020] [Indexed: 11/20/2022] Open
Abstract
Hemophagocytic syndromes comprise a cluster of hyperinflammatory disorders, including hemophagocytic lymphohistiocytosis and macrophage activation syndrome. Overwhelming macrophage activation has long been considered a final common pathway in the pathophysiology of hemophagocytic syndromes leading to the characteristic cytokine storm, laboratory abnormalities, and organ injuries that define the clinical spectrum of the disease. So far, it is unknown whether primary macrophage activation alone can induce the disease phenotype. In this study, we established a novel mouse model of a hemophagocytic syndrome by treating mice with an agonistic anti-CD40 antibody (Ab). The response in wild-type mice is characterized by a cytokine storm, associated with hyperferritinemia, high soluble CD25, erythrophagocytosis, secondary endothelial activation with multiple organ vaso-occlusion, necrotizing hepatitis, and variable cytopenias. The disease is dependent on a tumor necrosis factor-α-interferon-γ-driven amplification loop. After macrophage depletion with clodronate liposomes or in mice with a macrophage-selective deletion of the CD40 gene (CD40flox/flox/LysMCre), the disease was abolished. These data provide a new preclinical model of a hemophagocytic syndrome and reinforce the key pathophysiological role of macrophages.
Collapse
|
4
|
Liu G, Baird AW, Parsons MJ, Fan K, Skerrett-Byrne DA, Nair PM, Makanyengo S, Chen J, Neal R, Goggins BJ, Tay H, Mathe A, Soh WS, Minahan K, Hansbro PM, Nixon B, McCaughan GW, Holtmann G, Colgan SP, Keely S. Platelet activating factor receptor acts to limit colitis-induced liver inflammation. FASEB J 2020; 34:7718-7732. [PMID: 32293760 DOI: 10.1096/fj.201901779r] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 12/31/2022]
Abstract
Liver inflammation is a common extraintestinal manifestation in inflammatory bowel disease (IBD), yet, the mechanisms driving gut-liver axis inflammation remain poorly understood. IBD leads to a breakdown in the integrity of the intestinal barrier causing an increase in portal and systemic gut-derived antigens, which challenge the liver. Here, we examined the role of platelet activating factor receptor (PAFR) in colitis-associated liver damage using dextran sulfate sodium (DSS) and anti-CD40-induced colitis models. Both DSS and anti-CD40 models exhibited liver inflammation associated with colitis. Colitis reduced global PAFR protein expression in mouse livers causing an exclusive re-localization of PAFR to the portal triad. The global decrease in liver PAFR was associated with increased sirtuin 1 while relocalized PAFR expression was limited to Kupffer cells (KCs) and co-localized with toll-like receptor 4. DSS activated the NLRP3-inflammasome and increased interleukin (IL)-1β in the liver. Antagonism of PAFR amplified the inflammasome response by increasing NLRP3, caspase-1, and IL-1β protein levels in the liver. LPS also increased NLRP3 response in human hepatocytes, however, overexpression of PAFR restored the levels of NLPR3 and caspase-1 proteins. Interestingly, KCs depletion also increased IL-1β protein in mouse liver after DSS challenge. These data suggest a protective role for PAFR-expressing KCs during colitis and that regulation of PAFR is important for gut-liver axis homeostasis.
Collapse
Affiliation(s)
- Gang Liu
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, NSW, Australia
| | - Alan W Baird
- UCD School of Veterinary Medicine, University College Dublin, Dublin, Ireland
| | - Marie J Parsons
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Kening Fan
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - David A Skerrett-Byrne
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Environmental and Life Sciences, Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Prema M Nair
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Samwel Makanyengo
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Jinbiao Chen
- Liver Injury and Cancer Program, Centenary Research Institute, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Rachel Neal
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Bridie J Goggins
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Hock Tay
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia.,Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, NSW, Australia
| | - Andrea Mathe
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Wai S Soh
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Kyra Minahan
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| | - Phil M Hansbro
- School of Life Science, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.,Centre for Inflammation, Centenary Institute, Camperdown, NSW, Australia
| | - Brett Nixon
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Environmental and Life Sciences, Priority Research Centre for Reproductive Science, The University of Newcastle, Callaghan, NSW, Australia
| | - Geoffrey W McCaughan
- Liver Injury and Cancer Program, Centenary Research Institute, Camperdown, NSW, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Gerald Holtmann
- Department of Gastroenterology and Hepatology, Princess Alexandra Hospital, Woolloongabba, QLD, Australia.,Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, Australia
| | - Sean P Colgan
- University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Simon Keely
- Priority Research Centre for Digestive Health and Neurogastroenterology, The University of Newcastle, Newcastle, NSW, Australia.,Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
5
|
Li J, Tan H, Zhou X, Zhang C, Jin H, Tian Y, Zhao X, Li X, Sun X, Duan M, Zhang D. The Protection of Midazolam Against Immune Mediated Liver Injury Induced by Lipopolysaccharide and Galactosamine in Mice. Front Pharmacol 2019; 9:1528. [PMID: 30670973 PMCID: PMC6331471 DOI: 10.3389/fphar.2018.01528] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/13/2018] [Indexed: 01/23/2023] Open
Abstract
Objectives: Liver macrophages agitated by Lipopolysaccharide (LPS) can enhance immuno-inflammatory responses in the liver which mediate liver injury and result in dysfunction. Midazolam has been reported to have inhibitory effects on activated immunity and escalated inflammation, however, what the effects of midazolam on the liver injury caused by excessive immuno-inflammatory response in sepsis, and what influence it will exert on inflamed liver macrophages need to be elucidated. Methods: In the present study, LPS and galactosamine-induced acute liver injury mice were used to observe the effect of midazolam in vivo. LPS-stimulated bone marrow cells were used to evaluate the influence of midazolam on monocytes in vitro. Results: Midazolam prevented liver tissue injury and decreased serum alanine transaminase (ALT) level in LPS plus galactosamine treated mice. Mechanistically, midazolam suppressed tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) produced by LPS stimulated liver macrophages in vivo and bone marrow monocytes in vitro, and reduced the expression of major histocompatibility complex class II (MHC II), cluster of differentiation 40 and 86 (CD40 and CD86) on the cell surface. These results could be reversed by PK-11195, a peripheral benzodiazepine receptor (PBR) blocker. Conclusion: Midazolam can prevent liver from LPS-induced immune mediated liver injury by inhibiting inflammation and immune activation in liver macrophages.
Collapse
Affiliation(s)
- Jian Li
- Department of Intensive Care Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Clinical Research Institute, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Hong Tan
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China.,Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaona Zhou
- Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China.,General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Chunpan Zhang
- Beijing Clinical Research Institute, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China.,Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hua Jin
- Beijing Clinical Research Institute, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China.,Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yue Tian
- Beijing Clinical Research Institute, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China.,Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xinyan Zhao
- Beijing Clinical Research Institute, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Xinmin Li
- Beijing Clinical Research Institute, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China.,Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xuelian Sun
- Beijing Clinical Research Institute, Beijing, China.,Department of Emergency Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Meili Duan
- Department of Intensive Care Unit, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Clinical Research Institute, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China
| | - Dong Zhang
- Beijing Clinical Research Institute, Beijing, China.,Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, China.,Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
6
|
Rehermann B, Thimme R. Insights From Antiviral Therapy Into Immune Responses to Hepatitis B and C Virus Infection. Gastroenterology 2019; 156:369-383. [PMID: 30267712 PMCID: PMC6340757 DOI: 10.1053/j.gastro.2018.08.061] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/05/2018] [Accepted: 08/21/2018] [Indexed: 12/17/2022]
Abstract
There are 257 million persons worldwide with chronic hepatitis B virus (HBV) infection, a leading causes of liver cancer. Almost all adults with acute HBV infection have a rapid immune response to the virus, resulting in life-long immunity, but there is no cure for individuals with chronic HBV infection, which they acquire during early life. The mechanisms that drive the progression of HBV through distinct clinical phases to end-stage liver disease are poorly understood. Likewise, it is not clear whether and how immune responses can be modulated to allow control and/or clearance of intrahepatic HBV DNA. We review the innate and adaptive immune responses to acute and chronic HBV infections and responses to antiviral therapy. Comparisons with hepatitis C virus infection provide insights into the reversibility of innate inflammatory responses and the potential for successful therapy to recover virus-specific memory immune responses.
Collapse
Affiliation(s)
- Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland.
| | - Robert Thimme
- Klinik für Innere Medizin II, University Hospital Freiburg, Faculty of Medicine, Hugstetter Straße 55, 79106 Freiburg, Germany
| |
Collapse
|
7
|
Barthels C, Ogrinc A, Steyer V, Meier S, Simon F, Wimmer M, Blutke A, Straub T, Zimber-Strobl U, Lutgens E, Marconi P, Ohnmacht C, Garzetti D, Stecher B, Brocker T. CD40-signalling abrogates induction of RORγt + Treg cells by intestinal CD103 + DCs and causes fatal colitis. Nat Commun 2017; 8:14715. [PMID: 28276457 PMCID: PMC5347138 DOI: 10.1038/ncomms14715] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 01/25/2017] [Indexed: 12/23/2022] Open
Abstract
Immune homeostasis in intestinal tissues depends on the generation of regulatory T (Treg) cells. CD103+ dendritic cells (DCs) acquire microbiota-derived material from the gut lumen for transport to draining lymph nodes and generation of receptor-related orphan γt+ (RORγt+) Helios−-induced Treg (iTreg) cells. Here we show CD40-signalling as a microbe-independent signal that can induce migration of CD103+ DCs from the lamina propria (LP) to the mesenteric lymph nodes. Transgenic mice with constitutive CD11c-specific CD40-signalling have reduced numbers of CD103+ DCs in LP and a low frequency of RORγt+Helios− iTreg cells, exacerbated inflammatory Th1/Th17 responses, high titres of microbiota-specific immunoglobulins, dysbiosis and fatal colitis, but no pathology is detected in other tissues. Our data demonstrate a CD40-dependent mechanism capable of abrogating iTreg cell induction by DCs, and suggest that the CD40L/CD40-signalling axis might be able to intervene in the generation of new iTreg cells in order to counter-regulate immune suppression to enhance immunity. CD103+ dendritic cells induce iTreg cells to maintain immune balance in the gut, but how CD40-signalling regulates this process is unclear. Here the authors show that mice with constitutive CD11c-specific CD40-signalling have altered CD103+ dendritic cell migration, reduced iTreg cell induction, and fatal colitis.
Collapse
Affiliation(s)
- Christian Barthels
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Ana Ogrinc
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Verena Steyer
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Stefanie Meier
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Ferdinand Simon
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| | - Maria Wimmer
- Center of Allergy Environment (ZAUM), Helmholtz Center and TU Munich, Neuherberg 85764, Germany
| | - Andreas Blutke
- Section of Animal Pathology, Department of Veterinary Clinical Sciences, LMU Munich, Munich 80539, Germany
| | - Tobias Straub
- Bioinformatics core unit, BMC, LMU Munich, Großhaderner Strasse 9, Planegg-Munich 82152, Germany
| | | | - Esther Lutgens
- Institut für Prophylaxe und Epidemiologie der Kreislaufkrankheiten, LMU Munich, Munich 80336, Germany.,Department of Medical Biochemistry, AMC, Amsterdam 1105AZ, The Netherlands
| | - Peggy Marconi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara 44121, Italy
| | - Caspar Ohnmacht
- Center of Allergy Environment (ZAUM), Helmholtz Center and TU Munich, Neuherberg 85764, Germany
| | - Debora Garzetti
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, German Center for Infection Research (DZIF), Partner Site Munich, LMU Munich, Munich 80336, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, German Center for Infection Research (DZIF), Partner Site Munich, LMU Munich, Munich 80336, Germany
| | - Thomas Brocker
- Institute for Immunology, LMU Munich, Großhaderner Strasse 9, Planegg-Martinsried 82152, Germany
| |
Collapse
|
8
|
CD40-TNF activation in mice induces extended sickness behavior syndrome co-incident with but not dependent on activation of the kynurenine pathway. Brain Behav Immun 2015; 50:125-140. [PMID: 26173174 DOI: 10.1016/j.bbi.2015.06.184] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 06/23/2015] [Accepted: 06/27/2015] [Indexed: 12/12/2022] Open
Abstract
The similarity between sickness behavior syndrome (SBS) in infection and autoimmune disorders and certain symptoms in major depressive disorder (MDD), and the high co-morbidity of autoimmune disorders and MDD, constitutes some of the major evidence for the immune-inflammation hypothesis of MDD. CD40 ligand-CD40 immune-activation is important in host response to infection and in development of autoimmunity. Mice given a single intra-peritoneal injection of CD40 agonist antibody (CD40AB) develop SBS for 2-3days characterized by weight loss and increased sleep, effects that are dependent on the cytokine, tumor necrosis factor (TNF). Here we report that CD40AB also induces behavioral effects that extend beyond acute SBS and co-occur with but are not mediated by kynurenine pathway activation and recovery. CD40AB led to decreased saccharin drinking (days 1-7) and decreased Pavlovian fear conditioning (days 5-6), and was without effect on physical fatigue (day 5). These behavioral effects co-occurred with increased plasma and brain levels of kynurenine and its metabolites (days 1-7/8). Co-injection of TNF blocker etanercept with CD40AB prevented each of SBS, reduced saccharin drinking, and kynurenine pathway activation in plasma and brain. Repeated oral administration of a selective indoleamine 2,3-dioxygenase (IDO) inhibitor blocked activation of the kynurenine pathway but was without effect on SBS and saccharin drinking. This study provides novel evidence that CD40-TNF activation induces deficits in saccharin drinking and Pavlovian fear learning and activates the kynurenine pathway, and that CD40-TNF activation of the kynurenine pathway is not necessary for induction of the acute or extended SBS effects.
Collapse
|
9
|
Richards JA, Bucsaiova M, Hesketh EE, Ventre C, Henderson NC, Simpson K, Bellamy COC, Howie SEM, Anderton SM, Hughes J, Wigmore SJ. Acute Liver Injury Is Independent of B Cells or Immunoglobulin M. PLoS One 2015; 10:e0138688. [PMID: 26406765 PMCID: PMC4583453 DOI: 10.1371/journal.pone.0138688] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 09/02/2015] [Indexed: 01/08/2023] Open
Abstract
Background & Aims Acute liver injury is a clinically important pathology and results in the release of Danger Associated Molecular Patterns, which initiate an immune response. Withdrawal of the injurious agent and curtailing any pathogenic secondary immune response may allow spontaneous resolution of injury. The role B cells and Immunoglobulin M (IgM) play in acute liver injury is largely unknown and it was proposed that B cells and/or IgM would play a significant role in its pathogenesis. Methods Tissue from 3 models of experimental liver injury (ischemia-reperfusion injury, concanavalin A hepatitis and paracetamol-induced liver injury) and patients transplanted following paracetamol overdose were stained for evidence of IgM deposition. Mice deficient in B cells (and IgM) were used to dissect out the role B cells and/or IgM played in the development or resolution of injury. Serum transfer into mice lacking IgM was used to establish the role IgM plays in injury. Results Significant deposition of IgM was seen in the explanted livers of patients transplanted following paracetamol overdose as well as in 3 experimental models of acute liver injury (ischemia-reperfusion injury, concanavalin A hepatitis and paracetamol-induced liver injury). Serum transfer into IgM-deficient mice failed to reconstitute injury (p = 0.66), despite successful engraftment of IgM. Mice deficient in both T and B cells (RAG1-/-) mice (p<0.001), but not B cell deficient (μMT) mice (p = 0.93), were significantly protected from injury. Further interrogation with T cell deficient (CD3εKO) mice confirmed that the T cell component is a key mediator of sterile liver injury. Mice deficient in B cells and IgM mice did not have a significant delay in resolution following acute liver injury. Discussion IgM deposition appears to be common feature of both human and murine sterile liver injury. However, neither IgM nor B cells, play a significant role in the development of or resolution from acute liver injury. T cells appear to be key mediators of injury. In conclusion, the therapeutic targeting of IgM or B cells (e.g. with Rituximab) would have limited benefit in protecting patients from acute liver injury.
Collapse
Affiliation(s)
- James A. Richards
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
- Clinical Surgery, The University of Edinburgh, Edinburgh, United Kingdom
- * E-mail:
| | - Martina Bucsaiova
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Emily E. Hesketh
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Chiara Ventre
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Neil C. Henderson
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
- Hepatology, Division of Health Sciences, School of Clinical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Kenneth Simpson
- Hepatology, Division of Health Sciences, School of Clinical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Christopher O. C. Bellamy
- The University of Edinburgh, Edinburgh, United Kingdom
- Department of Pathology, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| | - Sarah E. M. Howie
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen M. Anderton
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jeremy Hughes
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen J. Wigmore
- MRC Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
- Clinical Surgery, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
10
|
Müller AF, Strauss L, Greter M, Gast H, Recher M, Becher B, Fontana A. Neutralization of colony-stimulating factor 1 receptor prevents sickness behavior syndrome by reprogramming inflammatory monocytes to produce IL-10. Brain Behav Immun 2015; 48:78-85. [PMID: 25749482 DOI: 10.1016/j.bbi.2015.02.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/18/2015] [Accepted: 02/25/2015] [Indexed: 10/23/2022] Open
Abstract
Sickness behavior syndrome (SBS) as characterized by fatigue and depression impairs quality of life in patients with inflammatory diseases caused by infections and autoimmunity. Systemic engagement of CD40 in mice leads to an inflammatory syndrome with acute hepatitis, lymphadenopathy and development of SBS as evidenced by induction of sleep and weight loss. In the study presented here we show that the elimination of resident tissue macrophages in mice by antibody-mediated neutralization of colony-stimulating factor-1 receptor (CSF1R) did not prevent CD40 induced hepatitis, but conferred resistance to the development of SBS. The protective effect of CSF1R mAb on weight loss and behavior changes induced by CD40 activation coincided with the transformation of pro-inflammatory monocytes to IL-10 producing myeloid cells. In IL-10 knockout mice CSF1R neutralization failed to exert protection from the occurrence of SBS. This study establishes the unexpected key role of CSF1R in the polarization of inflammatory monocytes and thereby SBS in inflammatory liver diseases.
Collapse
Affiliation(s)
| | - Laura Strauss
- Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Heidemarie Gast
- Department of Neurology, Inselspital, University Hospital Berne, Switzerland
| | - Mike Recher
- Immunodeficiency Clinic, Department Biomedicine, University Hospital Basel, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Switzerland
| | - Adriano Fontana
- Institute of Experimental Immunology, University of Zurich, Switzerland.
| |
Collapse
|
11
|
Thapa M, Chinnadurai R, Velazquez VM, Tedesco D, Elrod E, Han JH, Sharma P, Ibegbu C, Gewirtz A, Anania F, Pulendran B, Suthar MS, Grakoui A. Liver fibrosis occurs through dysregulation of MyD88-dependent innate B-cell activity. Hepatology 2015; 61:2067-79. [PMID: 25711908 PMCID: PMC4441566 DOI: 10.1002/hep.27761] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 02/23/2015] [Indexed: 12/18/2022]
Abstract
UNLABELLED Chronic liver disease mediated by activation of hepatic stellate cells (HSCs) leads to liver fibrosis. Here, we postulated that the immune regulatory properties of HSCs might promote the profibrogenic activity of B cells. Fibrosis is completely attenuated in carbon tetrachloride-treated, B cell-deficient µMT mice, showing that B cells are required. The retinoic acid produced by HSCs augmented B-cell survival, plasma cell marker CD138 expression, and immunoglobulin G production. These activities were reversed following addition of the retinoic acid inhibitor LE540. Transcriptional profiling of fibrotic liver B cells revealed increased expression of genes related to activation of nuclear factor κ light chain enhancer of activated B cells, proinflammatory cytokine production, and CD40 signaling, suggesting that these B cells are activated and may be acting as inflammatory cells. Biological validation experiments also revealed increased activation (CD44 and CD86 expression), constitutive immunoglobulin G production, and secretion of the proinflammatory cytokines tumor necrosis factor-α, monocyte chemoattractant protein-1, and macrophage inflammatory protein-1α. Likewise, targeted deletion of B-cell-intrinsic myeloid differentiation primary response gene 88 signaling, an innate adaptor with involvement in retinoic acid signaling, resulted in reduced infiltration of migratory CD11c(+) dendritic cells and Ly6C(++) monocytes and, hence, reduced liver pathology. CONCLUSION Liver fibrosis occurs through a mechanism of HSC-mediated augmentation of innate B-cell activity. These findings highlight B cells as important "first responders" of the intrahepatic immune environment.
Collapse
Affiliation(s)
- Manoj Thapa
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Raghavan Chinnadurai
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Victoria M. Velazquez
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Dana Tedesco
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Elizabeth Elrod
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Jin-Hwan Han
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Prachi Sharma
- Division of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322
| | - Chris Ibegbu
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322
| | - Andrew Gewirtz
- Department of Biology, Georgia State University, Atlanta, GA 30303
| | - Frank Anania
- Division of Digestive Diseases, Emory University School of Medicine, Atlanta, GA 30322
| | - Bali Pulendran
- Department of Pathology, Emory University School of Medicine, Atlanta, GA 30322
| | - Mehul S. Suthar
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322,Department of Pediatrics and Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322
| | - Arash Grakoui
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA 30322,Division of Infectious diseases, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
12
|
Shen HH, Bai BK, Wang YQ, Zhou GDE, Hou J, Hu Y, Zhao JM, Li BS, Huang HL, Mao PY. Serum soluble CD40 is associated with liver injury in patients with chronic hepatitis B. Exp Ther Med 2015; 9:999-1005. [PMID: 25667667 PMCID: PMC4316966 DOI: 10.3892/etm.2015.2182] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 11/17/2014] [Indexed: 01/17/2023] Open
Abstract
Soluble cluster of differentiation 40 (sCD40) is proteolytically cleaved from membrane-bound CD40 and binds to CD154, thereby inhibiting CD40-CD154-mediated immune responses. The aim of the present study was to clarify the role of sCD40 in chronic hepatitis B (CHB). The sCD40 levels in sera from 132 patients with CHB and 33 healthy individuals were retrospectively measured. sCD40 concentrations in patients with CHB were higher than those in healthy controls, and sCD40 levels correlated positively with serum levels of the liver dysfunction biomarkers alanine transaminase (ALT) and aspartate transaminase (AST). sCD40 concentrations increased with a rise in the severity of liver necroinflammation and fibrosis. Patients with >75% liver tissue staining positive for hepatitis B virus (HBV) antigen expression showed significantly lower sCD40 levels than those who stained negative for the HBV antigen. The area under the receiver operating characteristic curve of sCD40 was greater than that of ALT and AST; thus, sCD40 levels have a high diagnostic accuracy for detecting severe liver inflammation in patients with CHB, and could serve as an immunological marker of hepatic tissue injury.
Collapse
Affiliation(s)
- Hong-Hui Shen
- Institute of Infectious Diseases, Beijing 302 Hospital, Beijing 100039, P.R. China
| | - Bing-Ke Bai
- Institute of Infectious Diseases, Beijing 302 Hospital, Beijing 100039, P.R. China
| | - Ya-Qing Wang
- Department of Gastroenterology, Beijing 305 Hospital, Beijing 100017, P.R. China
| | - Guang-DE Zhou
- Department of Pathology, Beijing 302 Hospital, Beijing 100039, P.R. China
| | - Jun Hou
- Institute of Infectious Diseases, Beijing 302 Hospital, Beijing 100039, P.R. China
| | - Yan Hu
- Institute of Infectious Diseases, Beijing 302 Hospital, Beijing 100039, P.R. China
| | - Jing-Min Zhao
- Department of Pathology, Beijing 302 Hospital, Beijing 100039, P.R. China
| | - Bao-Sen Li
- Institute of Infectious Diseases, Beijing 302 Hospital, Beijing 100039, P.R. China
| | - Hai-Li Huang
- Department of Gastroenterology, General Hospital of PLA, Beijing 100853, P.R. China
| | - Pan-Yong Mao
- Institute of Infectious Diseases, Beijing 302 Hospital, Beijing 100039, P.R. China
| |
Collapse
|
13
|
STING agonists induce an innate antiviral immune response against hepatitis B virus. Antimicrob Agents Chemother 2014; 59:1273-81. [PMID: 25512416 DOI: 10.1128/aac.04321-14] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chronicity of hepatitis B virus (HBV) infection is due to the failure of a host to mount a sufficient immune response to clear the virus. The aim of this study was to identify small-molecular agonists of the pattern recognition receptor (PRR)-mediated innate immune response to control HBV infection. To achieve this goal, a coupled mouse macrophage and hepatocyte culture system mimicking the intrahepatic environment was established and used to screen small-molecular compounds that activate macrophages to produce cytokines, which in turn suppress HBV replication in a hepatocyte-derived stable cell line supporting HBV replication in a tetracycline-inducible manner. An agonist of the mouse stimulator of interferon (IFN) genes (STING), 5,6-dimethylxanthenone-4-acetic acid (DMXAA), was found to induce a robust cytokine response in macrophages that efficiently suppressed HBV replication in mouse hepatocytes by reducing the amount of cytoplasmic viral nucleocapsids. Profiling of cytokines induced by DMXAA and agonists of representative Toll-like receptors (TLRs) in mouse macrophages revealed that, unlike TLR agonists that induced a predominant inflammatory cytokine/chemokine response, the STING agonist induced a cytokine response dominated by type I IFNs. Moreover, as demonstrated in an HBV hydrodynamic mouse model, intraperitoneal administration of DMXAA significantly induced the expression of IFN-stimulated genes and reduced HBV DNA replication intermediates in the livers of mice. This study thus proves the concept that activation of the STING pathway induces an antiviral cytokine response against HBV and that the development of small-molecular human STING agonists as immunotherapeutic agents for treatment of chronic hepatitis B is warranted.
Collapse
|
14
|
Quiniou C, Domínguez-Punaro M, Cloutier F, Erfani A, Ennaciri J, Sivanesan D, Sanchez M, Chognard G, Hou X, Rivera JC, Beauchamp C, Charron G, Vilquin M, Kuchroo V, Michnick S, Rioux JD, Lesage S, Chemtob S. Specific targeting of the IL-23 receptor, using a novel small peptide noncompetitive antagonist, decreases the inflammatory response. Am J Physiol Regul Integr Comp Physiol 2014; 307:R1216-30. [DOI: 10.1152/ajpregu.00540.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
IL-23 is part of the IL-12 family of cytokines and is composed of the p19 subunit specific to IL-23 and the p40 subunit shared with IL-12. IL-23 specifically contributes to the inflammatory process of multiple chronic inflammatory autoimmune disorders, including psoriasis, multiple sclerosis, inflammatory bowel disease, and rheumatoid arthritis. So far, one antibody targeting the shared p40 subunit of IL-12 and IL-23, Ustekinumab, is approved clinically to treat psoriasis. However, there are no treatments inhibiting specifically the IL-23 proinflammatory response. We have developed small IL-23R-specific antagonists by designing all D-peptides arising from flexible regions of IL-23R. Of these peptides, we selected 2305 (teeeqqly), since in addition to its soluble properties, it inhibited IL-23-induced STAT3 phosphorylation in spleen cells. Peptide 2305 specifically binds to IL-23R/IL-12Rβ1-expressing HEK-293 cells and not to cells devoid of the receptor. Peptide 2305 showed functional selectivity by modulating IL-23-induced gene expression in IL-23R/IL-12Rβ1-expressing cells and in Jurkat cells; 2305 does not inhibit IL-12-induced cytokine expression in IL-12Rβ-IL-12Rβ2-HEK-293 cells. Finally, compared with anti-p40 treatment, 2305 effectively and selectively inhibits IL-23-induced inflammation in three in vivo mouse models: IL-23-induced ear inflammation, anti-CD40-induced systemic inflammatory response, and collagen-induced arthritis. We, hereby, describe the discovery and characterization of a potent IL-23R small-peptide modulator, 2305 (teeeqqly), that is effective in vivo. 2305 may be more convenient, less cumbersome, less costly, and most importantly, more specific than current biologics for the treatment of inflammatory conditions, and conceivably complement the actual therapies for these chronic and debilitating inflammatory diseases.
Collapse
Affiliation(s)
- Christiane Quiniou
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montréal, Canada
| | | | - Frank Cloutier
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montréal, Canada
| | - Atefeh Erfani
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montréal, Canada
| | - Jamila Ennaciri
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montréal, Canada
| | - Durgajini Sivanesan
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Mélanie Sanchez
- Department of Biochemistry, Université de Montréal, Montréal, Canada
| | - Gaëlle Chognard
- Maisonneuve-Rosemont Hospital, Research Center, Montreal, Canada
- Department of Microbiology and Immunology, Université de Montréal, Montréal, Canada
| | - Xin Hou
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montréal, Canada
| | | | | | | | - Marie Vilquin
- Maisonneuve-Rosemont Hospital, Research Center, Montreal, Canada
| | - Vijay Kuchroo
- Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; and
| | - Stephen Michnick
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - John D. Rioux
- Montreal Heart Institute, Montréal, Canada
- Department of Medicine, Université de Montréal, Montréal, Canada
| | - Sylvie Lesage
- Maisonneuve-Rosemont Hospital, Research Center, Montreal, Canada
- Department of Microbiology and Immunology, Université de Montréal, Montréal, Canada
| | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, Centre Hospitalier Universitaire Sainte-Justine Research Centre, Montréal, Canada
| |
Collapse
|
15
|
Gast H, Müller A, Lopez M, Meier D, Huber R, Dechent F, Prinz M, Emmenegger Y, Franken P, Birchler T, Fontana A. CD40 activation induces NREM sleep and modulates genes associated with sleep homeostasis. Brain Behav Immun 2013; 27:133-44. [PMID: 23072727 DOI: 10.1016/j.bbi.2012.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Revised: 10/02/2012] [Accepted: 10/02/2012] [Indexed: 01/26/2023] Open
Abstract
The T-cell derived cytokine CD40 ligand is overexpressed in patients with autoimmune diseases. Through activation of its receptor, CD40 ligand leads to a tumor necrosis factor (TNF) receptor 1 (TNFR1) dependent impairment of locomotor activity in mice. Here we report that this effect is explained through a promotion of sleep, which was specific to non-rapid eye movement (NREM) sleep while REM sleep was suppressed. The increase in NREM sleep was accompanied by a decrease in EEG delta power during NREM sleep and by a decrease in the expression of transcripts in the cerebral cortex known to be associated with homeostatic sleep drive, such as Homer1a, Early growth response 2, Neuronal pentraxin 2, and Fos-like antigen 2. The effect of CD40 activation was mimicked by peripheral TNF injection and prevented by the TNF blocker etanercept. Our study indicates that sleep-wake dysregulation in autoimmune diseases may result from CD40 induced TNF:TNFR1 mediated alterations of molecular pathways, which regulate sleep-wake behavior.
Collapse
Affiliation(s)
- Heidemarie Gast
- Department of Neurology, Inselspital, University Hospital Berne, University of Bern, Bern, Switzerland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
BiVax: a peptide/poly-IC subunit vaccine that mimics an acute infection elicits vast and effective anti-tumor CD8 T-cell responses. Cancer Immunol Immunother 2012; 62:787-99. [PMID: 23266830 DOI: 10.1007/s00262-012-1382-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 11/27/2012] [Indexed: 10/27/2022]
Abstract
Therapeutic vaccines for the treatment of cancer are an attractive alternative to some of the conventional therapies that are currently used. More importantly, vaccines could be very useful to prevent recurrences when applied after primary therapy. Unfortunately, most therapeutic vaccines for cancer have performed poorly due to the low level of immune responses that they induce. Previous work done in our laboratory in cancer mouse models demonstrated that vaccines consisting of synthetic peptides representing minimal CD8 T-cell epitopes administered i.v. mixed with poly-IC and anti-CD40 antibodies (TriVax) were capable of inducing massive T cell responses similar to those found during acute infections. We now report that some peptides are capable of inducing similarly large T cell responses after vaccination with poly-IC alone (BiVax). The results show that amphiphilic peptides are more likely to function as strong immunogens in BiVax and that systemic immunizations (i.v. or i.m.) were more effective than local (s.c.) vaccine administration. The immune responses induced by BiVax were found to be effective against established tumors in two mouse cancer models. The roles of various immune-related pathways such as type-I IFN, CD40 costimulation, CD4 T cells, TLRs and the MDA5 RNA helicase were examined. The present findings could facilitate the development of simple and effective subunit vaccines for diseases where CD8 T cells provide a therapeutic benefit.
Collapse
|
17
|
Moon JJ, Huang B, Irvine DJ. Engineering nano- and microparticles to tune immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2012; 24:3724-46. [PMID: 22641380 PMCID: PMC3786137 DOI: 10.1002/adma.201200446] [Citation(s) in RCA: 297] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Indexed: 05/13/2023]
Abstract
The immune system can be a cure or cause of disease, fulfilling a protective role in attacking cancer or pathogenic microbes but also causing tissue destruction in autoimmune disorders. Thus, therapies aimed to amplify or suppress immune reactions are of great interest. However, the complex regulation of the immune system, coupled with the potential systemic side effects associated with traditional systemic drug therapies, has presented a major hurdle for the development of successful immunotherapies. Recent progress in the design of synthetic micro- and nano-particles that can target drugs, deliver imaging agents, or stimulate immune cells directly through their physical and chemical properties is leading to new approaches to deliver vaccines, promote immune responses against tumors, and suppress autoimmunity. In addition, novel strategies, such as the use of particle-laden immune cells as living targeting agents for drugs, are providing exciting new approaches for immunotherapy. This progress report describes recent advances in the design of micro- and nano-particles for immunotherapies and diagnostics.
Collapse
Affiliation(s)
- James J Moon
- Dept. of Materials Science and Eng., Massachusetts Institute of Technology-MIT, Cambridge, MA, USA
| | | | | |
Collapse
|
18
|
Kwong B, Liu H, Irvine DJ. Induction of potent anti-tumor responses while eliminating systemic side effects via liposome-anchored combinatorial immunotherapy. Biomaterials 2011; 32:5134-47. [PMID: 21514665 PMCID: PMC3140866 DOI: 10.1016/j.biomaterials.2011.03.067] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 03/28/2011] [Indexed: 11/24/2022]
Abstract
Immunostimulatory therapies that activate immune response pathways are of great interest for overcoming the immunosuppression present in advanced tumors. Agonistic anti-CD40 antibodies and CpG oligonucleotides have previously demonstrated potent, synergistic anti-tumor effects, but their clinical use even as monotherapies is hampered by dose-limiting inflammatory toxicity provoked upon systemic exposure. We hypothesized that by anchoring immuno-agonist compounds to lipid nanoparticles we could retain the bioactivity of therapeutics in the local tumor tissue and tumor-draining lymph node, but limit systemic exposure to these potent molecules. We prepared PEGylated liposomes bearing surface-conjugated anti-CD40 and CpG and assessed their therapeutic efficacy and systemic toxicity compared to soluble versions of the same immuno-agonists, injected intratumorally in the B16F10 murine model of melanoma. Anti-CD40/CpG-liposomes significantly inhibited tumor growth and induced a survival benefit similar to locally injected soluble anti-CD40 + CpG. Biodistribution analyses following local delivery showed that the liposomal carriers successfully sequestered anti-CD40 and CpG in vivo, reducing leakage into systemic circulation while allowing draining to the tumor-proximal lymph node. Contrary to locally-administered soluble immunotherapy, anti-CD40/CpG-liposomes did not elicit significant increases in serum levels of ALT enzyme, systemic inflammatory cytokines, or overall weight loss, confirming that off-target inflammatory effects had been minimized. The development of a delivery strategy capable of inducing robust anti-tumor responses concurrent with minimal systemic side effects is crucial for the continued progress of potent immunotherapies toward widespread clinical translation.
Collapse
Affiliation(s)
- Brandon Kwong
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Mass. Ave., Cambridge, MA 02139, USA
| | | | | |
Collapse
|
19
|
Kimura K, Sekiguchi S, Hayashi S, Hayashi Y, Hishima T, Nagaki M, Kohara M. Role of interleukin-18 in intrahepatic inflammatory cell recruitment in acute liver injury. J Leukoc Biol 2010; 89:433-442. [DOI: 10.1189/jlb.0710412] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Abstract
ABSTRACT
Although the innate immune system has been demonstrated to play important roles as the first line of defense against various infections, little is known about the interactions between intrahepatic inflammatory cells and the cytokine network in the liver. Here, we examined the role of IL-18 in IHL recruitment in acute liver injury. C57BL/6 mice were injected with an αCD40 mAb, and their serum IL-18 levels were observed to increase, with subsequent recruitment of IHLs into the liver. NKT cells were involved in this liver injury, as the serum ALT levels were reduced in NKT KO mice through the suppression of macrophage and monocyte migration and cytokine production. In contrast, depletion of neutrophils exacerbated the liver injury associated with high levels of TNF-α and IL-18 and increased numbers of macrophages and monocytes. Treatment with a neutralizing antibody against IL-18 reduced the serum ALT levels and inflammatory cell accumulation in the liver. Finally, additional administration of rIL-18 with αCD40 injection caused severe liver injury with increased IFN-γ production by NK cells. In conclusion, these findings demonstrate that IL-18 modulates liver inflammation by the recruitment of inflammatory cells, including NKT cells, macrophages, monocytes, and neutrophils.
Collapse
Affiliation(s)
- Kiminori Kimura
- Division of Hepatology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital , Tokyo, Japan
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science , Tokyo, Japan
| | - Satoshi Sekiguchi
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science , Tokyo, Japan
| | - Seishu Hayashi
- Division of Hepatology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital , Tokyo, Japan
| | - Yukiko Hayashi
- Division of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital , Tokyo, Japan
| | - Tsunekazu Hishima
- Division of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital , Tokyo, Japan
| | - Masahito Nagaki
- Department of Gastroenterology, Gifu University Graduate School of Medicine , Gifu, Japan
| | - Michinori Kohara
- Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science , Tokyo, Japan
| |
Collapse
|
20
|
Kimura K, Nagaki M, Matsuura T, Moriwaki H, Kakimi K. Pathological role of CD44 on NKT cells in carbon tetrachloride-mediated liver injury. Hepatol Res 2009; 39:93-105. [PMID: 18721153 DOI: 10.1111/j.1872-034x.2008.00409.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM CD44 has a variety of functions in immune regulation and signal transduction. Although CD44 is involved in the induction of several inflammatory diseases, it remains unknown whether CD44-targeting therapies are useful for liver diseases. Here, we examined whether CD44 blockade is effective in a chemical-induced liver injury model. METHODS We injected CD44 knock out (KO) or wild type mice with carbon tetrachloride (CCl(4)) and examined the difference of liver injury by immunological or histological analysis. RESULTS Although CD44KO mice exhibited suppressed liver injury at 6 h after CCl(4) injection with decreased inflammatory cell numbers and cytokine production, these mice showed severe liver injury at 24 h. We found that NKT cells played an important role in liver injury with increased infiltration of theliver after migration, which was independent of the CD44 pathway. In CD44NKT double-KO mice, liver injury was suppressed with reduced cytokine production and macrophage infiltration compared with CD44KO mice. Furthermore, MIP-2 derived from NKT cells or tumor necrosis factor alpha from macrophages contributed to exacerbation of the liver injury, since neutralization of MIP-2 provided significant protection against liver injury in CD44KO mice. Finally, we found that CD44KO mice exhibited excessive liver fibrosis compared with wild-type mice after repeated CCl(4) injections. CONCLUSION We found that CD44 has unique characteristics for inflammatory liver diseases associated with NKT cell infiltration and activation. Furthermore, CD44-targeting therapies may need to be viewed with caution for liver diseases due to the actions of the liver immune system.
Collapse
Affiliation(s)
- Kiminori Kimura
- Division of Hepatology, Tokyo Metropolitan Cancer and Infectious diseases Center Komagome Hospital, 18-22-3, Honkomagome, Bunkyoku, Tokyo 113-8677, Japan
| | | | | | | | | |
Collapse
|
21
|
Law CL, Grewal IS. Therapeutic interventions targeting CD40L (CD154) and CD40: the opportunities and challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:8-36. [PMID: 19760064 DOI: 10.1007/978-0-387-89520-8_2] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CD40 was originally identified as a receptor on B-cells that delivers contact-dependent T helper signals to B-cells through interaction with CD40 ligand (CD40L, CD154). The pivotal role played by CD40-CD40L interaction is illustrated by the defects in B-lineage cell development and the altered structures of secondary lymphoid tissues in patients and engineered mice deficient in CD40 or CD40L. CD40 signaling also provides critical functions in stimulating antigen presentation, priming of helper and cytotoxic T-cells and a variety of inflammatory reactions. As such, dysregulations in the CD40-CD40L costimulation pathway are prominently featured in human diseases ranging from inflammatory conditions to systemic autoimmunity and tissue-specific autoimmune diseases. Moreover, studies in CD40-expressing cancers have provided convincing evidence that the CD40-CD40L pathway regulates survival of neoplastic cells as well as presentation of tumor-associated antigens to the immune system. Extensive research has been devoted to explore CD40 and CD40L as drug targets. A number of anti-CD40L and anti-CD40 antibodies with diverse biological effects are in clinical development for treatment of cancer and autoimmune diseases. This chapter reviews the role of CD40-CD40L costimulation in disease pathogenesis, the characteristics of therapeutic agents targeting this pathway and status of their clinical development.
Collapse
Affiliation(s)
- Che-Leung Law
- Department of Preclinical Therapeutics, Seattle Genetics Inc., 21823 30th Drive SE, Bothell, Washington, 98021, USA.
| | | |
Collapse
|
22
|
Hamzah J, Nelson D, Moldenhauer G, Arnold B, Hämmerling GJ, Ganss R. Vascular targeting of anti-CD40 antibodies and IL-2 into autochthonous tumors enhances immunotherapy in mice. J Clin Invest 2008; 118:1691-9. [PMID: 18398504 DOI: 10.1172/jci33201] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2007] [Accepted: 02/20/2008] [Indexed: 11/17/2022] Open
Abstract
Current anticancer therapy is a delicate balance between elimination of malignant cells and harmful side effects for the host. In this study, we used a tumor-homing peptide to engineer anti-CD40 agonist antibodies and recombinant IL-2 such that they were selectively delivered into spontaneously arising tumors in a transgenic mouse model of islet cell carcinogenesis. Intravenous injection of these agents, either separately or together, led to accumulation in the vicinity of tumor neovessels without toxic side effects. Although both molecules are critical for adaptive immunity, the most profound effects were seen in endothelial cells. Combined, local anti-CD40 and IL-2 therapy reduced tumor vascularity and significantly delayed tumor growth in mice. Remarkably, tumor-bearing mice remained disease-free long-term when targeted anti-CD40 and IL-2 were combined with transfers of preactivated antitumor immune cells. In this therapeutic setting, triggering of CD40 on endothelial cells induced an inflammatory response of the vessel wall and facilitated effector cell accumulation in the tumor parenchyma while IL-2 promoted antigen-specific immune cell persistence. We believe this is a novel and highly effective anticancer approach, whereby tumor stroma is "conditioned" for enhanced immune cell entry and survival, facilitating immune-mediated tumor destruction and leading to a sustained antitumor response.
Collapse
Affiliation(s)
- Juliana Hamzah
- Western Australian Institute for Medical Research, University of Western Australia Centre for Medical Research, Perth, Western Australia, Australia
| | | | | | | | | | | |
Collapse
|
23
|
Kimura K, Nagaki M, Kakimi K, Saio M, Saeki T, Okuda Y, Kuwata K, Moriwaki H. Critical role of CD44 in hepatotoxin-mediated liver injury. J Hepatol 2008; 48:952-61. [PMID: 18395288 DOI: 10.1016/j.jhep.2008.01.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 12/05/2007] [Accepted: 01/03/2008] [Indexed: 01/27/2023]
Abstract
BACKGROUND/AIMS Blocking of adhesion molecules is considered to be one of the therapeutic strategies inflammatory diseases, although it remains unclear whether this strategy is beneficial. METHODS We used CD44-deficient mice to assess whether inhibition of CD44 could control liver injury caused by carbon tetrachloride (CCl(4)). RESULTS CD44-deficient mice exhibited suppressed liver inflammation during the early phase (within 6h) after CCl(4) injection due to reduced inflammatory cell infiltration and cytokine production, but showed severe liver inflammation with increased numbers of apoptotic hepatocytes at the late phase (after 12h). The induction of hepatocyte apoptosis was triggered by reduced NF-kappaB activity, which was induced by the low inflammatory cytokine concentrations. Furthermore, macrophages contributed to the induction of hepatocyte apoptosis, since neutralization by an anti-CD11b antibody significantly protected against hepatocyte apoptosis. Finally, we found that blocking of MIP-2 and TNF-alpha reduced hepatocyte apoptosis with decreased numbers of intrahepatic leukocytes and reduced inflammatory cytokine production. CONCLUSIONS These findings suggest that targeting of CD44 as a therapeutic approach for inflammatory liver diseases may require caution for particular immune systems in the liver.
Collapse
Affiliation(s)
- Kiminori Kimura
- Department of Immunotherapeutics (Medinet), Graduate School of Medicine, The University of Tokyo, 1-3-7 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Enhanced efficacy and reduced toxicity of multifactorial adjuvants compared with unitary adjuvants as cancer vaccines. Blood 2008; 111:3116-25. [PMID: 18202224 DOI: 10.1182/blood-2007-09-114371] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Identification of Toll-like receptors (TLRs) and their ligands, and tumor necrosis factor-tumor necrosis factor receptor (TNF-TNFR) pairs have provided the first logical, hypothesis-based strategies to molecularly concoct adjuvants to elicit potent cell-mediated immunity via activation of innate and adaptive immunity. However, isolated activation of one immune pathway in the absence of others can be toxic, ineffective, and detrimental to long-term, protective immunity. Effective engineered vaccines must include agents that trigger multiple immunologic pathways. Here, we report that combinatorial use of CD40 and TLR agonists as a cancer vaccine, compared with monotherapy, elicits high frequencies of self-reactive CD8(+) T cells, potent tumor-specific CD8(+) memory, CD8(+) T cells that efficiently infiltrate the tumor-burdened target organ; therapeutic efficacy; heightened ratios of CD8(+) T cells to FoxP3(+) cells at the tumor site; and reduced hepatotoxicity. These findings provide intelligent strategies for the formulation of multifactorial vaccines to achieve maximal efficacy in cancer vaccine trials in humans.
Collapse
|
25
|
Ogiso T, Nagaki M, Takai S, Tsukada Y, Mukai T, Kimura K, Moriwaki H. Granulocyte colony-stimulating factor impairs liver regeneration in mice through the up-regulation of interleukin-1beta. J Hepatol 2007; 47:816-25. [PMID: 17869372 DOI: 10.1016/j.jhep.2007.06.017] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Revised: 05/25/2007] [Accepted: 06/14/2007] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIMS Stem cell induction via granulocyte colony-stimulating factor (G-CSF) administration is utilized in the treatment of various diseases. Therefore, we examined the effect of G-CSF administration to a liver fibrosis model induced by dimethylnitrosamine (DMN). METHODS ICR mice were subcutaneously injected with either G-CSF (150microg/kg) or saline at days 0, 3, 7 and 10. Subacute liver injury was established by intraperitoneal injection of DMN (10mg/kg) on three consecutive days of each week. RESULTS G-CSF administration significantly decreased the survival rate of mice treated with DMN. There was no difference in the degree of liver injury or fibrosis between either group of mice. However, assessment by proliferating cell nuclear antigen (PCNA) revealed that the G-CSF-treated mice experienced a greater degree of inhibition of liver cell proliferation than the control mice. Interleukin-1beta (IL-1beta) mRNA expression increased in the livers of G-CSF-treated mice. PCNA staining and analysis of cell cycle-related proteins also revealed that passive immunization with anti-IL-1beta-neutralizing antibody improved the impaired hepatocellular regeneration and resulted in an improved survival rate of mice treated with G-CSF and DMN. CONCLUSIONS G-CSF administration suppressed liver cell proliferation through the up-regulation of IL-1beta expression in DMN-induced liver injury.
Collapse
Affiliation(s)
- Tomio Ogiso
- Department of Gastroenterology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Yu J, Hui AY, Chu ESH, Cheng ASL, Go MYY, Chan HLY, Leung WK, Cheung KF, Ching AKK, Chui YL, Chan KK, Sung JJY. Expression of a cyclo-oxygenase-2 transgene in murine liver causes hepatitis. Gut 2007; 56:991-9. [PMID: 17148503 PMCID: PMC1994375 DOI: 10.1136/gut.2006.097923] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND It has been proved that cyclo-oxygenase-2 (COX-2) is rapidly induced by inflammatory mediators. However, it is not known whether overexpression of COX-2 in the liver is sufficient to promote activation or secretion of inflammatory factors leading to hepatitis. AIM To investigate the role forced expression of COX-2 in liver by using inducible COX-2 transgenic (TG) mice. METHODS TG mice that overexpress the human COX-2 gene in the liver using the liver-specific transthyretin promoter and non-TG littermates were derived and fed the normal diet for up to 12 months. Hepatic prostaglandin E(2) (PGE(2)) content was determined using enzyme immunoassay, nuclear factor kappaB (NF-kappaB) activation by electrophoretic mobility shift assays, apoptosis by terminal deoxynucleotidyl transferase-mediated dUTP-digoxigenin nick end labelling and proliferation by Ki-67 immunohistochemistry. RESULTS COX-2 TG mice exhibited strongly increased COX-2 and PGE(2), elevated serum alanine aminotransferase level and histological hepatitis. Hepatic COX-2 expression in the TG mice resulted in activation of NF-kappaB and inflammatory cytokine cascade, with a marked expression of the proinflammatory cytokines tumour necrosis factor (TNF)-alpha (9.4-fold), interleukin (IL)-6 (4.4-fold), IL-1beta (3.6-fold), and of the anti-inflammatory cytokine IL-10 (4.4-fold) and chemokine macrophage inflammatory protein-2 (3.2-fold). The inflammatory response of the COX-2 TG mice was associated with infiltration macrophages and lymphocytes, increased cell proliferation and high rates of cell apoptosis. Administration of the COX-2 inhibitor celecoxib in TG mice restored liver histology to normal. CONCLUSION Enhanced COX-2 expression in hepatocytes is sufficient to induce hepatitis by activating NF-kappaB, stimulating the secretion of proinflammatory cytokines, recruiting macrophage and altering cell kinetics. Inhibition of COX-2 represents a mechanism-based chemopreventive approach to hepatitis.
Collapse
Affiliation(s)
- Jun Yu
- Department of Medicine and Therapeutics, Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Bartholdy C, Kauffmann SØ, Christensen JP, Thomsen AR. Agonistic anti-CD40 antibody profoundly suppresses the immune response to infection with lymphocytic choriomeningitis virus. THE JOURNAL OF IMMUNOLOGY 2007; 178:1662-70. [PMID: 17237416 DOI: 10.4049/jimmunol.178.3.1662] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous work has shown that agonistic Abs to CD40 (anti-CD40) can boost weak CD8 T cell responses as well as substitute for CD4 T cell function during chronic gammaherpes virus infection. Agonistic anti-CD40 treatment has, therefore, been suggested as a potential therapeutic strategy in immunocompromised patients. In this study, we investigated whether agonistic anti-CD40 could substitute for CD4 T cell help in generating a sustained CD8 T cell response and prevent viral recrudescence following infection with lymphocytic choriomeningitis virus (LCMV). Contrary to expectations, we found that anti-CD40 treatment of MHC class II-deficient mice infected with a moderate dose of LCMV resulted in severe suppression of the antiviral CD8 T cell response and uncontrolled virus spread, rather than improved CD8 T cell immune surveillance. In Ab-treated wild-type mice, the antiviral CD8 T cell response also collapsed prematurely, and virus clearance was delayed. Additional analysis revealed that, following anti-CD40 treatment, the virus-specific CD8 T cells initially proliferated normally, but an increased cell loss compared with that in untreated mice was observed. The anti-CD40-induced abortion of virus-specific CD8 T cells during LCMV infection was IL-12 independent, but depended partly on Fas expression. Notably, similar anti-CD40 treatment of vesicular stomatitis virus-infected mice resulted in an improved antiviral CD8 T cell response, demonstrating that the effect of anti-CD40 treatment varies with the virus infection studied. For this reason, we recommend further evaluation of the safety of this regimen before being applied to human patients.
Collapse
Affiliation(s)
- Christina Bartholdy
- Institute of Medical Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | |
Collapse
|