1
|
Suzuki K, Tajima M, Tokumaru Y, Oshiro Y, Nagata S, Kamada H, Kihara M, Nakano K, Honjo T, Ohta A. Anti-PD-1 antibodies recognizing the membrane-proximal region are PD-1 agonists that can down-regulate inflammatory diseases. Sci Immunol 2023; 8:eadd4947. [PMID: 36638191 DOI: 10.1126/sciimmunol.add4947] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The PD-1 receptor triggers a negative immunoregulatory mechanism that prevents overactivation of immune cells and subsequent inflammatory diseases. Because of its biological significance, PD-1 has been a drug target for modulating immune responses. Immunoenhancing anti-PD-1 blocking antibodies have become a widely used cancer treatment; however, little is known about the required characteristics for anti-PD-1 antibodies to be capable of stimulating immunosuppressive activity. Here, we show that PD-1 agonists exist in the group of anti-PD-1 antibodies recognizing the membrane-proximal extracellular region in sharp contrast to the binding of the membrane-distal region by blocking antibodies. This trend was consistent in an analysis of 81 anti-human PD-1 monoclonal antibodies. Because PD-1 agonist antibodies trigger immunosuppressive signaling by cross-linking PD-1 molecules, Fc engineering to enhance FcγRIIB binding of PD-1 agonist antibodies notably improved human T cell inhibition. A PD-1 agonist antibody suppressed inflammation in murine disease models, indicating its clinical potential for treatment of various inflammatory disorders, including autoimmune diseases.
Collapse
Affiliation(s)
- Kensuke Suzuki
- Department of Immunology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan.,Pharmaceutical R&D Division, Meiji Seika Pharma Co. Ltd., Tokyo 104-8002, Japan
| | - Masaki Tajima
- Department of Immunology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan.,Division of Integrated High-Order Regulatory Systems, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yosuke Tokumaru
- Department of Immunology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan.,Pharmaceutical R&D Division, Meiji Seika Pharma Co. Ltd., Tokyo 104-8002, Japan
| | - Yuya Oshiro
- Department of Immunology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan.,Pharmaceutical R&D Division, Meiji Seika Pharma Co. Ltd., Tokyo 104-8002, Japan
| | - Satoshi Nagata
- Laboratory of Antibody Design, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health, and Nutrition, Ibaraki 567-0085, Japan
| | - Haruhiko Kamada
- Laboratory of Antibody Design, Center for Drug Design Research, National Institutes of Biomedical Innovation, Health, and Nutrition, Ibaraki 567-0085, Japan
| | - Miho Kihara
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Kohei Nakano
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe 650-0047, Japan
| | - Tasuku Honjo
- Department of Immunology and Genomic Medicine, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Akio Ohta
- Department of Immunology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe 650-0047, Japan
| |
Collapse
|
2
|
Multiparametric Flow Cytometry-Based Immunophenotyping of Mouse Liver Immune Cells. Methods Protoc 2022; 5:mps5050070. [PMID: 36136816 PMCID: PMC9498390 DOI: 10.3390/mps5050070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
The liver is a complex organ that governs many types of metabolisms, including energy metabolism and other cellular processes. The liver also plays a crucial role in important functions in immunity, and the activity of liver tissue-associated immunity affects the outcome of many liver pathologies. A thorough characterization of the liver immune microenvironment may contribute to a better understanding of immune signaling, the mechanisms of specific immune responses, and even to improved predictions about therapy outcomes. In this paper, we present an optimized, simple, and rapid protocol to characterize the liver-associated immune cell milieu. We believe that the most suitable technique for obtaining a complex immune cell suspension and for removing contaminating blood cells is to perform mouse liver perfusion, using only phosphate buffer saline. Combining an enzymatic digestion and a mechanical dissociation of liver tissue, followed by cell purification, improves downstream applications. This combination is an essential prerequisite for immune cell determination and characterization. We then demonstrate a flow cytometry-based multiparametric immunophenotyping along with a gating strategy to detect and quantify liver endothelial cells, T cells (helper and cytotoxic), B cells, NK cells, NKT cells, neutrophils, monocytes (subsets included), dendritic cells (subsets included), macrophages and Kupffer cells.
Collapse
|
3
|
Lopes ME, Nakagaki BN, Mattos MS, Campolina-Silva GH, Meira RDO, Paixão P, Oliveira A, Faustino L, Gonçalves R, Menezes GB. Susceptibility to Infections During Acute Liver Injury Depends on Transient Disruption of Liver Macrophage Niche. Front Immunol 2022; 13:892114. [PMID: 35967353 PMCID: PMC9368782 DOI: 10.3389/fimmu.2022.892114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Kupffer cells are the primary liver resident immune cell responsible for the liver firewall function, including clearance of bacterial infection from the circulation, as they are strategically positioned inside the liver sinusoid with intimate contact with the blood. Disruption in the tissue-resident macrophage niche, such as in Kupffer cells, can lead to a window of susceptibility to systemic infections, which represents a significant cause of mortality in patients with acetaminophen (APAP) overdose-induced acute liver injury (ALI). However, how Kupffer cell niche disruption increases susceptibility to systemic infections in ALI is not fully understood. Using a mouse model of ALI induced by APAP overdose, we found that Kupffer cells upregulated the apoptotic cell death program and were markedly reduced in the necrotic areas during the early stages of ALI, opening the niche for the infiltration of neutrophils and monocyte subsets. In addition, during the resolution phase of ALI, the remaining tissue macrophages with a Kupffer cell morphology were observed forming replicating cell clusters closer to necrotic areas devoid of Kupffer cells. Interestingly, mice with APAP-induced liver injury were still susceptible to infections despite the dual cellular input of circulating monocytes and proliferation of remaining Kupffer cells in the damaged liver. Therapy with bone marrow-derived macrophages (BMDM) was shown to be effective in occupying the niche devoid of Kupffer cells following APAP-induced ALI. The rapid BMDM migration to the liver and their positioning within necrotic areas enhanced the healing of the tissue and restored the liver firewall function after BMDM therapy. Therefore, we showed that disruption in the Kupffer cell niche and its impaired function during acute liver injury are key factors for the susceptibility to systemic bacterial infections. In addition, modulation of the liver macrophage niche was shown to be a promising therapeutic strategy for liver injuries that reduce the Kupffer cell number and compromise the organ function.
Collapse
Affiliation(s)
- Mateus Eustáquio Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- *Correspondence: Mateus Eustáquio Lopes, ; Gustavo Batista Menezes,
| | - Brenda Naemi Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Matheus Silvério Mattos
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gabriel Henrique Campolina-Silva
- Department of Biochemistry and Immunology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Québec, QC, Canada
| | - Raquel de Oliveira Meira
- Macrophage and Monocyte Biology Laboratory, Department of Pathology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Pierre Henrique de Menezes Paixão
- Macrophage and Monocyte Biology Laboratory, Department of Pathology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - André Gustavo Oliveira
- Department of Physiology and Biophysics, Instituto de Ciências Biológicas, Universidade
Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Lucas D. Faustino
- Center for Immunology and Inflammatory Diseases, Division of Rheumatology, Allergy and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Ricardo Gonçalves
- Macrophage and Monocyte Biology Laboratory, Department of Pathology, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
- *Correspondence: Mateus Eustáquio Lopes, ; Gustavo Batista Menezes,
| |
Collapse
|
4
|
Abstract
Mass cytometry has revolutionized immunophenotyping, particularly in exploratory settings where simultaneous breadth and depth of characterization of immune populations is needed with limited samples such as in preclinical and clinical tumor immunotherapy. Mass cytometry is also a powerful tool for single-cell immunological assays, especially for complex and simultaneous characterization of diverse intratumoral immune subsets or immunotherapeutic cell populations. Through the elimination of spectral overlap seen in optical flow cytometry by replacement of fluorescent labels with metal isotopes, mass cytometry allows, on average, robust analysis of 60 individual parameters simultaneously. This is, however, associated with significantly increased complexity in the design, execution, and interpretation of mass cytometry experiments. To address the key pitfalls associated with the fragmentation, complexity, and analysis of data in mass cytometry for immunologists who are novices to these techniques, we have developed a comprehensive resource guide. Included in this review are experiment and panel design, antibody conjugations, sample staining, sample acquisition, and data pre-processing and analysis. Where feasible multiple resources for the same process are compared, allowing researchers experienced in flow cytometry but with minimal mass cytometry expertise to develop a data-driven and streamlined project workflow. It is our hope that this manuscript will prove a useful resource for both beginning and advanced users of mass cytometry.
Collapse
Affiliation(s)
- Akshay Iyer
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anouk A. J. Hamers
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Asha B. Pillai
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Sheila and David Fuente Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
5
|
Moreira TG, Mangani D, Cox LM, Leibowitz J, Lobo ELC, Oliveira MA, Gauthier CD, Nakagaki BN, Willocq V, Song A, Guo L, Lima DCA, Murugaiyan G, Butovsky O, Gabriely G, Anderson AC, Rezende RM, Faria AMC, Weiner HL. PD-L1 + and XCR1 + dendritic cells are region-specific regulators of gut homeostasis. Nat Commun 2021; 12:4907. [PMID: 34389726 PMCID: PMC8363668 DOI: 10.1038/s41467-021-25115-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 07/26/2021] [Indexed: 12/12/2022] Open
Abstract
The intestinal mucosa constitutes an environment of closely regulated immune cells. Dendritic cells (DC) interact with the gut microbiome and antigens and are important in maintaining gut homeostasis. Here, we investigate DC transcriptome, phenotype and function in five anatomical locations of the gut lamina propria (LP) which constitute different antigenic environments. We show that DC from distinct gut LP compartments induce distinct T cell differentiation and cytokine secretion. We also find that PD-L1+ DC in the duodenal LP and XCR1+ DC in the colonic LP comprise distinct tolerogenic DC subsets that are crucial for gut homeostasis. Mice lacking PD-L1+ and XCR1+ DC have a proinflammatory gut milieu associated with an increase in Th1/Th17 cells and a decrease in Treg cells and have exacerbated disease in the models of 5-FU-induced mucositis and DSS-induced colitis. Our findings identify PD-L1+ and XCR1+ DC as region-specific physiologic regulators of intestinal homeostasis.
Collapse
Affiliation(s)
- Thais G Moreira
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Davide Mangani
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura M Cox
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeffrey Leibowitz
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eduardo L C Lobo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mariana A Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Christian D Gauthier
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Brenda N Nakagaki
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Valerie Willocq
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anya Song
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lydia Guo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - David C A Lima
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gopal Murugaiyan
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oleg Butovsky
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Galina Gabriely
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana C Anderson
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rafael M Rezende
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana Maria C Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Howard L Weiner
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Xiao Z, Liu L, Jin Y, Pei X, Sun W, Wang M. A Potential Prophylactic Probiotic for Inflammatory Bowel Disease: The Overall Investigation of Clostridium tyrobutyricum ATCC25755 Attenuates LPS-Induced Inflammation via Regulating Intestinal Immune Cells. Mol Nutr Food Res 2021; 65:e2001213. [PMID: 34021704 DOI: 10.1002/mnfr.202001213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/29/2021] [Indexed: 12/14/2022]
Abstract
SCOPE This study aims to roundly investigate whether Clostridium tyrobutyricum (Ct) alleviates inflammation via regulating immune cells in the small intestines. METHODS AND RESULTS Mice are pre-treated with different concentrations of Ct follow by LPS stimulation. Ct maintains the mice body weight under inflammation. In response to LPS, 107 CFU mL-1 Ct decreases the mRNA expression of inflammatory cytokines in the duodenum, while 108 CFU mL-1 Ct reduces inflammatory cytokines expression in both duodenum and ileum and protected intestinal morphology. The small intestinal immune cells are analyzed using flow cytometry. Ct decreases the numbers of macrophages and mast cells in the intestines in response to LPS. In the duodenum, Ct enhances dentritic cells (DCs), regulatory T cells (Tregs), and T helper cell 17 (Th17) proportions. Ct decreases DCs and Tregs proportions, while enhances Th17 numbers in the ileum. The underlying mechanism of Ct in preventing inflammation may rely on the physiological immune cell composition of the intestines. In response to LPS, Ct may mainly stimulate Tregs via activating DCs in the duodenum while trigger Th17 cells in the ileum, thereby maintaining the intestinal homeostasis. CONCLUSION Ct alleviates the LPS-induce inflammation via regulating different immune cell types in the small intestines, highlighting that Ct is a potential prophylactic probiotic in intestinal diseases.
Collapse
Affiliation(s)
- Zhiping Xiao
- The key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Lujie Liu
- The key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Yuyue Jin
- The key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Xun Pei
- The key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Wanjing Sun
- The key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Minqi Wang
- The key Laboratory of Molecular Animal Nutrition, Ministry of Education, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
7
|
Garo LP, Ajay AK, Fujiwara M, Gabriely G, Raheja R, Kuhn C, Kenyon B, Skillin N, Kadowaki-Saga R, Saxena S, Murugaiyan G. MicroRNA-146a limits tumorigenic inflammation in colorectal cancer. Nat Commun 2021; 12:2419. [PMID: 33893298 PMCID: PMC8065171 DOI: 10.1038/s41467-021-22641-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation can drive tumor development. Here, we have identified microRNA-146a (miR-146a) as a major negative regulator of colonic inflammation and associated tumorigenesis by modulating IL-17 responses. MiR-146a-deficient mice are susceptible to both colitis-associated and sporadic colorectal cancer (CRC), presenting with enhanced tumorigenic IL-17 signaling. Within myeloid cells, miR-146a targets RIPK2, a NOD2 signaling intermediate, to limit myeloid cell-derived IL-17-inducing cytokines and restrict colonic IL-17. Accordingly, myeloid-specific miR-146a deletion promotes CRC. Moreover, within intestinal epithelial cells (IECs), miR-146a targets TRAF6, an IL-17R signaling intermediate, to restrict IEC responsiveness to IL-17. MiR-146a within IECs further suppresses CRC by targeting PTGES2, a PGE2 synthesis enzyme. IEC-specific miR-146a deletion therefore promotes CRC. Importantly, preclinical administration of miR-146a mimic, or small molecule inhibition of the miR-146a targets, TRAF6 and RIPK2, ameliorates colonic inflammation and CRC. MiR-146a overexpression or miR-146a target inhibition represent therapeutic approaches that limit pathways converging on tumorigenic IL-17 signaling in CRC.
Collapse
Affiliation(s)
- Lucien P Garo
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Boston University School of Medicine, Boston, MA, USA
| | - Amrendra K Ajay
- Renal Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Mai Fujiwara
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Galina Gabriely
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Radhika Raheja
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Chantal Kuhn
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Brendan Kenyon
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Nathaniel Skillin
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ryoko Kadowaki-Saga
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Shrishti Saxena
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gopal Murugaiyan
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Programing of an Intravascular Immune Firewall by the Gut Microbiota Protects against Pathogen Dissemination during Infection. Cell Host Microbe 2020; 28:660-668.e4. [PMID: 32810440 DOI: 10.1016/j.chom.2020.07.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/09/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
Eradication of pathogens from the bloodstream is critical to prevent disseminated infections and sepsis. Kupffer cells in the liver form an intravascular firewall that captures and clears pathogens from the blood. Here, we show that the catching and killing of circulating pathogens by Kupffer cells in vivo are promoted by the gut microbiota through commensal-derived D-lactate that reaches the liver via the portal vein. The integrity of this Kupffer cell-mediated intravascular firewall requires continuous crosstalk with gut commensals, as microbiota depletion with antibiotics leads to a failure of pathogen clearance and overwhelming disseminated infection. Furthermore, administration of purified D-lactate to germ-free mice, or gnotobiotic colonization with D-lactate-producing commensals, restores Kupffer cell-mediated pathogen clearance by the liver firewall. Thus, the gut microbiota programs an intravascular immune firewall that protects against the spread of bacterial infections via the bloodstream.
Collapse
|
9
|
Diniz AB, Antunes MM, Lacerda VADS, Nakagaki BN, Freitas Lopes MA, Castro-Oliveira HMD, Mattos MS, Mafra K, de Miranda CDM, de Oliveira Costa KM, Lopes ME, Alvarenga DM, Carvalho-Gontijo R, Marchesi SC, Lacerda DR, de Araújo AM, de Carvalho É, David BA, Santos MM, Lima CX, Silva Gomes JA, Minto Fontes Cal TC, de Souza BR, Couto CA, Faria LC, Teixeira Vidigal PV, Matos Ferreira AV, Radhakrishnnan S, Ricci M, Oliveira AG, Rezende RM, Menezes GB. Imaging and immunometabolic phenotyping uncover changes in the hepatic immune response in the early phases of NAFLD. JHEP Rep 2020; 2:100117. [PMID: 32695965 PMCID: PMC7365949 DOI: 10.1016/j.jhepr.2020.100117] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/02/2020] [Accepted: 03/26/2020] [Indexed: 12/25/2022] Open
Abstract
Background & Aims The precise determination of non-alcoholic fatty liver disease (NAFLD) onset is challenging. Thus, the initial hepatic responses to fat accumulation, which may be fundamental to our understanding of NAFLD evolution and clinical outcomes, are largely unknown. Herein, we chronologically mapped the immunologic and metabolic changes in the liver during the early stages of fatty liver disease in mice and compared this with human NAFLD samples. Methods Liver biopsies from patients with NAFLD (NAFLD activity score [NAS] 2–3) were collected for gene expression profiling. Mice received a high-fat diet for short periods to mimic initial steatosis and the hepatic immune response was investigated using a combination of confocal intravital imaging, gene expression, cell isolation, flow cytometry and bone marrow transplantation assays. Results We observed major immunologic changes in patients with NAS 2–3 and in mice in the initial stages of NAFLD. In mice, these changes significantly increased mortality rates upon drug-induced liver injury, as well as predisposing mice to bacterial infections. Moreover, deletion of Toll-like receptor 4 in liver cells dampened tolerogenesis, particularly in Kupffer cells, in the initial stages of dietary insult. Conclusion The hepatic immune system acts as a sentinel for early and minor changes in hepatic lipid content, mounting a biphasic response upon dietary insult. Priming of liver immune cells by gut-derived Toll-like receptor 4 ligands plays an important role in liver tolerance in initial phases, but continuous exposure to insults may lead to damage and reduced ability to control infections. Lay summary Fatty liver is a very common form of hepatic disease, leading to millions of cases of cirrhosis every year. Patients are often asymptomatic until becoming very sick. Therefore, it is important that we expand our knowledge of the early stages of disease pathogenesis, to enable early diagnosis. Herein, we show that even in the early stages of fatty liver disease, there are significant alterations in genes involved in the inflammatory response, suggesting that the hepatic immune system is disturbed even following minor and undetectable changes in liver fat content. This could have implications for the diagnosis and clinical management of fatty liver disease. Hepatic immune response is already altered in liver biopsies from patients with mild NAFLD. We designed a novel mouse model to mimic mild NAFLD, enabling the chronological mapping of liver changes. This revealed an increased mortality rate upon secondary liver damage and a window of increased susceptibility to infection. NAFLD diagnosis may be significantly improved by a more profound investigation of changes in hepatic immunology. These data could guide customized nutritional and therapeutic interventions at different stages of NAFLD.
Collapse
Key Words
- ALT, alanine aminotransferase
- APAP, acetaminophen
- CFUs, colony forming units
- DCs, dendritic cells
- E. coli, Escherichia coli
- HFD, high-fat diet
- ITT, insulin tolerance test
- KCs, Kupffer cells
- NAFLD
- NAFLD, non-alcoholic fatty liver disease
- NAS, NAFLD activity score
- NPCs, non-parenchymal cells
- SD, standard diet
- TLR4, Toll-like receptor 4
- WT, wild-type
- diet
- immune system
- immunity
- in vivo imaging
- liver
- metabolism
- steatosis
Collapse
Affiliation(s)
- Ariane Barros Diniz
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Viviane Aparecida de Souza Lacerda
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Brenda Naemi Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Maria Alice Freitas Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Hortência Maciel de Castro-Oliveira
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Matheus Silvério Mattos
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Kassiana Mafra
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Camila Dutra Moreira de Miranda
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Karen Marques de Oliveira Costa
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Mateus Eustáquio Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Débora Moreira Alvarenga
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | - Sarah Cozzer Marchesi
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | - Alan Moreira de Araújo
- Department of Pharmacodynamics, University of Florida, College of Pharmacy, Gainesville, FL, USA
| | - Érika de Carvalho
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | - Mônica Morais Santos
- Laboratório de Morfologia, Departamento de Biologia Animal, Universidade Federal de Viçosa, Viçosa, Brazil
| | - Cristiano Xavier Lima
- Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | | | - Bruna Roque de Souza
- Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Cláudia Alves Couto
- Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Luciana Costa Faria
- Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | | | | | | | | | | | - Rafael Machado Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
- Corresponding author. Address: Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antonio Carlos, 6627 - Belo Horizonte, Minas Gerais, 31270-901, Brazil. Tel./fax: +5531 3409 3015.
| |
Collapse
|
10
|
Garo LP, Ajay AK, Fujiwara M, Beynon V, Kuhn C, Gabriely G, Sadhukan S, Raheja R, Rubino S, Weiner HL, Murugaiyan G. Smad7 Controls Immunoregulatory PDL2/1-PD1 Signaling in Intestinal Inflammation and Autoimmunity. Cell Rep 2019; 28:3353-3366.e5. [PMID: 31553906 PMCID: PMC6925592 DOI: 10.1016/j.celrep.2019.07.065] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 06/09/2019] [Accepted: 07/18/2019] [Indexed: 02/08/2023] Open
Abstract
Smad7, a negative regulator of TGF-β signaling, has been implicated in the pathogenesis and treatment of inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC). Here, we found that Smad7 mediates intestinal inflammation by limiting the PDL2/1-PD1 axis in dendritic cells (DCs) and CD4+T cells. Smad7 deficiency in DCs promotes TGF-β responsiveness and the co-inhibitory molecules PDL2/1 on DCs, and it further imprints T cell-PD1 signaling to promote Treg differentiation. DC-specific Smad7 deletion mitigates DSS-induced colitis by inducing CD103+PDL2/1+DCs and Tregs. In addition, Smad7 deficiency in CD4+T cells promotes PD1 and PD1-induced Tregs in vitro. The transfer of Smad7-deficient CD4+T cells enhances Tregs in vivo and protects against T cell-mediated colitis. Furthermore, Smad7 antisense ameliorates DSS-induced UC, increasing TGF-β and PDL2/1-PD1 signaling. Enhancing PD1 signaling directly via Fc-fused PDL2/1 is also beneficial. Our results identify how Smad7 mediates intestinal inflammation and leverages these pathways therapeutically, providing additional strategies for IBD intervention.
Collapse
Affiliation(s)
- Lucien P Garo
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Amrendra K Ajay
- Renal Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mai Fujiwara
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Vanessa Beynon
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Chantal Kuhn
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Galina Gabriely
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Supriya Sadhukan
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Radhika Raheja
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stephen Rubino
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Gopal Murugaiyan
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Takenaka MC, Gabriely G, Rothhammer V, Mascanfroni ID, Wheeler MA, Chao CC, Gutiérrez-Vázquez C, Kenison J, Tjon EC, Barroso A, Vandeventer T, de Lima KA, Rothweiler S, Mayo L, Ghannam S, Zandee S, Healy L, Sherr D, Farez MF, Prat A, Antel J, Reardon DA, Zhang H, Robson SC, Getz G, Weiner HL, Quintana FJ. Control of tumor-associated macrophages and T cells in glioblastoma via AHR and CD39. Nat Neurosci 2019; 22:729-740. [PMID: 30962630 PMCID: PMC8052632 DOI: 10.1038/s41593-019-0370-y] [Citation(s) in RCA: 330] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 02/20/2019] [Indexed: 01/07/2023]
Abstract
Tumor-associated macrophages (TAMs) play an important role in the immune response to cancer, but the mechanisms by which the tumor microenvironment controls TAMs and T cell immunity are not completely understood. Here we report that kynurenine produced by glioblastoma cells activates aryl hydrocarbon receptor (AHR) in TAMs to modulate their function and T cell immunity. AHR promotes CCR2 expression, driving TAM recruitment in response to CCL2. AHR also drives the expression of KLF4 and suppresses NF-κB activation in TAMs. Finally, AHR drives the expression of the ectonucleotidase CD39 in TAMs, which promotes CD8+ T cell dysfunction by producing adenosine in cooperation with CD73. In humans, the expression of AHR and CD39 was highest in grade 4 glioma, and high AHR expression was associated with poor prognosis. In summary, AHR and CD39 expressed in TAMs participate in the regulation of the immune response in glioblastoma and constitute potential targets for immunotherapy.
Collapse
Affiliation(s)
- Maisa C Takenaka
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Galina Gabriely
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ivan D Mascanfroni
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Chun-Cheih Chao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina Gutiérrez-Vázquez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jessica Kenison
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily C Tjon
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andreia Barroso
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tyler Vandeventer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kalil Alves de Lima
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sonja Rothweiler
- Divisions of Gastroenterology, Hepatology and Transplantation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Lior Mayo
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Soufiene Ghannam
- Neuroimmunology Research Lab., Center for Excellence in Neuromics, Department of Neuroscience, Université de Montréal, Montréal, Québec, Canada
| | - Stephanie Zandee
- Neuroimmunology Research Lab., Center for Excellence in Neuromics, Department of Neuroscience, Université de Montréal, Montréal, Québec, Canada
| | - Luke Healy
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| | - David Sherr
- Dept of Environmental Health, Boston University School of Public Health, Boston, MA, USA
| | - Mauricio F Farez
- Center for Research on Neuroimmunological Diseases (CIEN), Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires, Argentina
- Center for Epidemiology, Biostatistics and Public Health (CEBES), Raúl Carrea Institute for Neurological Research (FLENI), Buenos Aires, Argentina
| | - Alexandre Prat
- Neuroimmunology Research Lab., Center for Excellence in Neuromics, Department of Neuroscience, Université de Montréal, Montréal, Québec, Canada
| | - Jack Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| | - David A Reardon
- Center for Neuro-Oncology, Dana Farber Cancer Institute, Brigham and Women's Hospital, Boston, MA, USA
| | - Hailei Zhang
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Simon C Robson
- Divisions of Gastroenterology, Hepatology and Transplantation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Gad Getz
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
12
|
Moreira TG, Horta LS, Gomes-Santos AC, Oliveira RP, Queiroz NMGP, Mangani D, Daniel B, Vieira AT, Liu S, Rodrigues AM, Gomes DA, Gabriely G, Ferreira E, Weiner HL, Rezende RM, Nagy L, Faria AMC. CLA-supplemented diet accelerates experimental colorectal cancer by inducing TGF-β-producing macrophages and T cells. Mucosal Immunol 2019; 12:188-199. [PMID: 30279515 DOI: 10.1038/s41385-018-0090-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/24/2018] [Accepted: 09/07/2018] [Indexed: 02/07/2023]
Abstract
Conjugated linoleic acid (CLA) has been shown to activate the nuclear receptor PPAR-γ and modulate metabolic and immune functions. Despite the worldwide use of CLA dietary supplementation, strong scientific evidence for its proposed beneficial actions are missing. We found that CLA-supplemented diet reduced mucosal damage and inflammatory infiltrate in the dextran sodium sulfate (DSS)-induced colitis model. Conditional deletion of PPAR-γ in macrophages from mice supplemented with CLA diet resulted in loss of this protective effect of CLA, suggesting a PPAR-γ-dependent mechanism mediated by macrophages. However, CLA supplementation significantly worsened colorectal tumor formation induced by azoxymethane and DSS by inducing macrophage and T-cell-producing TGF-β via PPAR-γ activation. Accordingly, either macrophage-specific deletion of PPAR-γ or in vivo neutralization of latency-associated peptide (LAP, a membrane-bound TGF-β)-expressing cells abrogated the protumorigenic effect of CLA. Thus, the anti-inflammatory properties of CLA are associated with prevention of colitis but also with development of colorectal cancer.
Collapse
Affiliation(s)
- T G Moreira
- Departamento de Alimentos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Campus Pampulha, 31270-901, Belo Horizonte, MG, Brazil. .,Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil. .,Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Biochemistry and Molecular Biology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary.
| | - L S Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - A C Gomes-Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - R P Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - N M G P Queiroz
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - D Mangani
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - B Daniel
- Department of Biochemistry and Molecular Biology, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary.,Department of Medicine, School of Medicine, Johns Hopkins All Children's Hospital, Johns Hopkins University, St. Petersburg, FL, 33701, USA
| | - A T Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - S Liu
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - A M Rodrigues
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - D A Gomes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - G Gabriely
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - E Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - H L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - R M Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - L Nagy
- Diabetes and Obesity Research Center, Sanford Burnham Medical Research Institute, Lake Nona, Orlando, FL, USA.,Department of Medicine, School of Medicine, Johns Hopkins All Children's Hospital, Johns Hopkins University, St. Petersburg, FL, 33701, USA
| | - A M C Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
13
|
Rubino SJ, Mayo L, Wimmer I, Siedler V, Brunner F, Hametner S, Madi A, Lanser A, Moreira T, Donnelly D, Cox L, Rezende RM, Butovsky O, Lassmann H, Weiner HL. Acute microglia ablation induces neurodegeneration in the somatosensory system. Nat Commun 2018; 9:4578. [PMID: 30385785 PMCID: PMC6212411 DOI: 10.1038/s41467-018-05929-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/29/2018] [Indexed: 12/24/2022] Open
Abstract
Previous studies have reported that microglia depletion leads to impairment of synapse formation and these cells rapidly repopulate from CNS progenitors. However, the impact of microglia depletion and repopulation in the long-term state of the CNS environment has not been characterized. Here, we report that acute and synchronous microglia depletion and subsequent repopulation induces gray matter microgliosis, neuronal death in the somatosensory cortex and ataxia-like behavior. We find a type 1 interferon inflammatory signature in degenerating somatosensory cortex from microglia-depleted mice. Transcriptomic and mass cytometry analysis of repopulated microglia demonstrates an interferon regulatory factor 7-driven activation state. Minocycline and anti-IFNAR1 antibody treatment attenuate the CNS type 1 interferon-driven inflammation, restore microglia homeostasis and reduce ataxic behavior. Neither microglia depletion nor repopulation impact neuropathology or T-cell responses during experimental autoimmune encephalomyelitis. Together, we found that acute microglia ablation induces a type 1 interferon activation state of gray matter microglia associated with acute neurodegeneration. Previous studies have shown that depletion of microglia at early developmental stages leads to neuronal death. Here the authors use an inducible system to ablate microglia in adulthood, showing that such depletion leads to ataxia-like behavior and neuronal loss, and identifying the inflammatory components that may contribute.
Collapse
Affiliation(s)
- Stephen J Rubino
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Lior Mayo
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA.,School of Molecular Cell Biology & Biotechnology, George S. Wise Faculty of Life Sciences, and Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, 69978, Israel
| | - Isabella Wimmer
- Department of Neuroimmunology, Medical University of Vienna, Spitalgasse 4, Vienna, 1090, Austria
| | - Victoria Siedler
- Department of Neuroimmunology, Medical University of Vienna, Spitalgasse 4, Vienna, 1090, Austria
| | - Florian Brunner
- Department of Neuroimmunology, Medical University of Vienna, Spitalgasse 4, Vienna, 1090, Austria
| | - Simon Hametner
- Department of Neuroimmunology, Medical University of Vienna, Spitalgasse 4, Vienna, 1090, Austria
| | - Asaf Madi
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA.,Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Amanda Lanser
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Thais Moreira
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Dustin Donnelly
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Laura Cox
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Rafael Machado Rezende
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Oleg Butovsky
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA.,Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA
| | - Hans Lassmann
- Department of Neuroimmunology, Medical University of Vienna, Spitalgasse 4, Vienna, 1090, Austria
| | - Howard L Weiner
- Ann Romney Center for Neurological Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA. .,Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, 02115, MA, USA.
| |
Collapse
|
14
|
Araujo AMD, Antunes MM, Mattos MS, Diniz AB, Alvarenga DM, Nakagaki BN, Carvalho ÉD, Lacerda VAS, Carvalho-Gontijo R, Goulart J, Mafra K, Freitas-Lopes MA, Oliveira HMDC, Dutra CM, David BA, Mendes Silva A, Quesniaux V, Ryffel B, Oliveira SC, Barber GN, Mansur DS, Cunha TM, Rezende RM, Oliveira AG, Menezes GB. Liver Immune Cells Release Type 1 Interferon Due to DNA Sensing and Amplify Liver Injury from Acetaminophen Overdose. Cells 2018; 7:cells7080088. [PMID: 30060463 PMCID: PMC6115735 DOI: 10.3390/cells7080088] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 01/07/2023] Open
Abstract
Hepatocytes may rupture after a drug overdose, and their intracellular contents act as damage-associated molecular patterns (DAMPs) that lead to additional leukocyte infiltration, amplifying the original injury. Necrosis-derived DNA can be recognized as a DAMP, activating liver non-parenchymal cells (NPCs). We hypothesized that NPCs react to DNA by releasing interferon (IFN)-1, which amplifies acetaminophen (APAP)-triggered liver necrosis. We orally overdosed different knockout mouse strains to investigate the pathways involved in DNA-mediated amplification of APAP-induced necrosis. Mice were imaged under intravital confocal microscopy to estimate injury progression, and hepatocytes and liver NPCs were differentially isolated for gene expression assays. Flow cytometry (FACS) using a fluorescent reporter mouse estimated the interferon-beta production by liver leukocytes under different injury conditions. We also treated mice with DNase to investigate the role of necrosis DNA signaling in IFN-1 production. Hepatocytes released a large amount of DNA after APAP overdose, which was not primarily sensed by these cells. However, liver NPCs promptly sensed such environmental disturbances and activated several DNA sensing pathways. Liver NPCs synthesized and released IFN-1, which was associated with concomitant hepatocyte necrosis. Ablation of IFN-1 recognition in interferon α/β receptor (IFNAR−/−) mice delayed APAP-mediated liver necrosis and dampened IFN-1 sensing pathways. We demonstrated a novel loop involving DNA recognition by hepatic NPCs and additional IFN-1 mediated hepatocyte death.
Collapse
Affiliation(s)
- Alan Moreira de Araujo
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Matheus Silvério Mattos
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Ariane Barros Diniz
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Débora Moreira Alvarenga
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Brenda Naemi Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Érika de Carvalho
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Viviane Aparecida Souza Lacerda
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Raquel Carvalho-Gontijo
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Jorge Goulart
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Kassiana Mafra
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Maria Alice Freitas-Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Hortência Maciel de Castro Oliveira
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Camila Miranda Dutra
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Bruna Araújo David
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Aristóbolo Mendes Silva
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Valerie Quesniaux
- Experimental and Molecular Immunology and Neurogenetics CNRS, University of Orleans, 45000 Orleans, France.
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and Neurogenetics CNRS, University of Orleans, 45000 Orleans, France.
| | - Sergio Costa Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Glen N Barber
- Department of Cell Biology and the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | - Daniel Santos Mansur
- Laboratory of Immunobiology, Universidade Federal de Santa Catarina, Santa Catarina 88040-900, Brazil.
| | - Thiago Mattar Cunha
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, São Paulo 14049-900, Brazil.
| | - Rafael Machado Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - André Gustavo Oliveira
- Departamento de Fisiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627-Belo Horizonte, Minas Gerais 31270-901, Brazil.
| |
Collapse
|
15
|
Antunes MM, Carvalho ÉD, Menezes GB. DIY: "Do Imaging Yourself" - Conventional microscopes as powerful tools for in vivo investigation. Int J Biochem Cell Biol 2017; 94:1-5. [PMID: 29128683 DOI: 10.1016/j.biocel.2017.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 01/29/2023]
Abstract
Intravital imaging has been increasingly employed in cell biology studies and it is becoming one of the most powerful tools for in vivo investigation. Although some protocols can be extremely complex, most intravital imaging procedures can be performed using basic surgery and animal maintenance techniques. More importantly, regular confocal microscopes - the same that are used for imaging immunofluorescence slides - can also acquire high quality intravital images and movies after minor adaptations. Here we propose minimal adaptations in stock microscopes that allow major improvements in different fields of scientific investigation.
Collapse
Affiliation(s)
- Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Érika de Carvalho
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
16
|
Nakagaki BN, Freitas-Lopes MA, Carvalho É, Carvalho-Gontijo R, Castro-Oliveira HM, Rezende RM, Cara DC, Santos MM, Lopes RP, David BA, Menezes GB. Generation of a triple-fluorescent mouse strain allows a dynamic and spatial visualization of different liver phagocytes in vivo. AN ACAD BRAS CIENC 2017; 91:e20170317. [PMID: 29044327 DOI: 10.1590/0001-3765201720170317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 06/28/2017] [Indexed: 12/31/2022] Open
Abstract
Resident and circulating immune cells have been extensively studied due to their almost ubiquitous role in cell biology. Despite their classification under the "immune cell department", it is becoming increasingly clear that these cells are involved in many different non-immune related phenomena, including fetus development, vascular formation, memory, social behavior and many other phenotypes. There is a huge potential in combining high-throughput assays - including flow cytometry and gene analysis - with in vivo imaging. This can improve our knowledge in both basic and clinical cell biology, and accessing the expression of markers that are relevant in the context of both homeostasis and disease conditions might be instrumental. Here we describe how we generated a novel mouse strain that spontaneously express three different fluorescence markers under control of well-studied receptors (CX3CR1, CCR2 and CD11c) that are involved in a plethora of stages of cell ontogenesis, maturation, migration and behavior. Also, we assess the percentage of the expression and co-expression of each marker under homeostasis conditions, and how these cells behave when a local inflammation is induced in the liver applying a cutting-edge technology to image cells by confocal intravital microscopy.
Collapse
Affiliation(s)
- Brenda N Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Maria A Freitas-Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Érika Carvalho
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Raquel Carvalho-Gontijo
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Hortência M Castro-Oliveira
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Rafael M Rezende
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, 02115, Boston, MA, United States of America
| | - Denise C Cara
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| | - Mônica M Santos
- Departamento de Biologia Animal, Universidade Federal de Viçosa, Av. Peter Henry Rolfs, s/n, Campus Universitário, 36570-900 Viçosa, MG, Brazil
| | - Rodrigo Pestana Lopes
- BD Biosciences, Rua Alexandre Dumas, 1976, Chácara Santo Antônio, 04717-040 São Paulo, SP, Brazil
| | - Bruna A David
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil.,Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo/USP, Av. Bandeirantes, 3900, Monte Alegre, 14049-900 Ribeirão Preto, SP, Brazil
| | - Gustavo B Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, sala N3-140, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Presidente Antônio Carlos, 6627, Pampulha, 31270-901 Belo Horizonte, MG, Brazil
| |
Collapse
|
17
|
Antunes MM, Araújo AM, Diniz AB, Pereira RVS, Alvarenga DM, David BA, Rocha RM, Lopes MAF, Marchesi SC, Nakagaki BN, Carvalho É, Marques PE, Ryffel B, Quesniaux V, Guabiraba Brito R, Filho JCA, Cara DC, Rezende RM, Menezes GB. IL-33 signalling in liver immune cells enhances drug-induced liver injury and inflammation. Inflamm Res 2017; 67:77-88. [PMID: 29032512 DOI: 10.1007/s00011-017-1098-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/15/2017] [Accepted: 09/27/2017] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE AND DESIGN The aim of this study was to investigate the contribution of IL-33/ST2 axis in the onset and progression of acute liver injury using a mice model of drug-induced liver injury (DILI). MATERIAL AND TREATMENTS DILI was induced by overdose administration of acetaminophen (APAP) by oral gavage in wild-type BALB/c, ST2-deficient mice and in different bone marrow chimeras. Neutrophils were depleted by anti-Ly6G and macrophages with clodronate liposomes (CLL). METHODS Blood and liver were collected for biochemical, immunologic and genetic analyses. Mice were imaged by confocal intravital microscopy and liver non-parenchymal cells and hepatocytes were isolated for flow cytometry, genetic and immunofluorescence studies. RESULTS Acetaminophen overdose caused a massive necrosis and accumulation of immune cells within the liver, concomitantly with IL-33 and chemokine release. Liver non-parenchymal cells were the major sensors for IL-33, and amongst them, neutrophils were the major players in amplification of the inflammatory response triggered by IL-33/ST2 signalling pathway. CONCLUSION Blockage of IL-33/ST2 axis reduces APAP-mediated organ injury by dampening liver chemokine release and activation of resident and infiltrating liver non-parenchymal cells.
Collapse
Affiliation(s)
- Maísa Mota Antunes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Alan Moreira Araújo
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Ariane Barros Diniz
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Rafaela Vaz Sousa Pereira
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Débora Moreira Alvarenga
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Bruna Araújo David
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Renata Monti Rocha
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Maria Alice Freitas Lopes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Sarah Cozzer Marchesi
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Brenda Naemi Nakagaki
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Érika Carvalho
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Pedro Elias Marques
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and Neurogenetics CNRS, University of Orleans, Orleans, France
| | - Valérie Quesniaux
- Experimental and Molecular Immunology and Neurogenetics CNRS, University of Orleans, Orleans, France
| | | | - José Carlos Alves Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Denise Carmona Cara
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Rafael Machado Rezende
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gustavo Batista Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| |
Collapse
|