1
|
Kinsella RL, Sur Chowdhury C, Smirnov A, Mreyoud Y, Kimmey JM, Esaulova E, McKee SR, Pride A, Kreamalmeyer D, Artyomov MN, Stallings CL. ATG5 suppresses type I IFN-dependent neutrophil effector functions during Mycobacterium tuberculosis infection in mice. Nat Microbiol 2025:10.1038/s41564-025-01988-8. [PMID: 40374743 DOI: 10.1038/s41564-025-01988-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 03/19/2025] [Indexed: 05/18/2025]
Abstract
Inflammation is critical for controlling infections but can cause disease when unchecked. During Mycobacterium tuberculosis (Mtb) infection, neutrophil-dominated inflammation is associated with exacerbated disease. ATG5 expression by neutrophils mediates autophagy-independent control of infection but mechanistic understanding of how this regulates protective neutrophil function is lacking. Using genetic mouse models along with in vivo and in vitro infection systems, we report herein that ATG5 is required in neutrophils to suppress type I interferon-induced PAD4-mediated histone citrullination and neutrophil extracellular trap (NET) release. In addition, ATG5 suppresses type I interferon-induced CXCL2 secretion and neutrophil swarming during Mtb infection. Elevated type I IFN signalling and NET release contribute to the early susceptibility of Atg5fl/fl-LysM-Cre mice during infection. These findings identify ATG5 as a master regulator of how type I interferon influences neutrophil responses during infection, revealing a potential target for host-directed therapies.
Collapse
Affiliation(s)
- Rachel L Kinsella
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA.
- Department of Medicine, Division of Infectious Diseases and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA.
| | - Chanchal Sur Chowdhury
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Asya Smirnov
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Yassin Mreyoud
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Jacqueline M Kimmey
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
- Department of Microbiology and Environmental Toxicology, UC Santa Cruz, Santa Cruz, CA, USA
| | - Ekaterina Esaulova
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Samuel R McKee
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Aaron Pride
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Darren Kreamalmeyer
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Christina L Stallings
- Department of Molecular Microbiology, Center for Women's Infectious Disease Research, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
2
|
Iliakis CS, Crotta S, Wack A. The Interplay Between Innate Immunity and Nonimmune Cells in Lung Damage, Inflammation, and Repair. Annu Rev Immunol 2025; 43:395-422. [PMID: 40036704 DOI: 10.1146/annurev-immunol-082323-031852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
As the site of gas exchange, the lung is critical for organismal survival. It is also subject to continual environmental insults inflicted by pathogens, particles, and toxins. Sometimes, these insults result in structural damage and the initiation of an innate immune response. Operating in parallel, the immune response aims to eliminate the threat, while the repair process ensures continual physiological function of the lung. The inflammatory response and repair processes are thus inextricably linked in time and space but are often studied in isolation. Here, we review the interplay of innate immune cells and nonimmune cells during lung insult and repair. We highlight how cellular cross talk can fine-tune the circuitry of the immune response, how innate immune cells can facilitate or antagonize proper organ repair, and the prolonged changes to lung immunity and physiology that can result from acute immune responses and repair processes.
Collapse
Affiliation(s)
- Chrysante S Iliakis
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Stefania Crotta
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| | - Andreas Wack
- Immunoregulation Laboratory, The Francis Crick Institute, London, United Kingdom;
| |
Collapse
|
3
|
Skeldon AM, Wang L, Sgarioto N, Beveridge RE, Chan S, Dorich S, Dumais V, Fradet N, Gaudreault S, LeGros P, McKay D, Seliniotakis R, Sietsema DV, Zhang L, Boily MO, Burch JD, Caron A, Fader LD, Lama L, Xie W, Patel DJ, Tuschl T, Crackower MA, Pike KA. Structural insight into the cGAS active site explains differences between therapeutically relevant species. Commun Chem 2025; 8:88. [PMID: 40121343 PMCID: PMC11929900 DOI: 10.1038/s42004-025-01481-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) is an intracellular sensor of double-stranded DNA that triggers a pro-inflammatory response upon binding. The interest in cGAS as a drug discovery target has increased substantially over the past decade due to growing evidence linking its activation to numerous peripheral and neurological diseases. Here, we report the binding mode of previously described cGAS inhibitors while also uncovering the structural basis for the interspecies potency shifts within this chemotype. A single threonine to isoleucine substitution between human and mouse cGAS drives compound activity, as demonstrated by biochemical, cellular, and in vivo studies. Finally, we utilize a structurally enabled design approach to engineer a novel chemical inhibitor with excellent potency for both human and mouse enzymes by targeting key interactions within the enzyme active site. Overall, this work provides the framework for rational optimization of cGAS inhibitors and potential preclinical translational strategies.
Collapse
Affiliation(s)
| | - Li Wang
- Ventus Therapeutics, Waltham, MA, USA
| | | | | | - Silas Chan
- Ventus Therapeutics, Saint-Laurent, QC, Canada
| | | | | | | | | | | | | | | | | | | | | | | | - Alex Caron
- Ventus Therapeutics, Saint-Laurent, QC, Canada
| | - Lee D Fader
- Ventus Therapeutics, Saint-Laurent, QC, Canada
| | - Lodoe Lama
- Laboratory for RNA Molecular Biology, The Rockefeller University, New York, NY, USA
| | - Wei Xie
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Dinshaw J Patel
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Thomas Tuschl
- Laboratory for RNA Molecular Biology, The Rockefeller University, New York, NY, USA
| | | | | |
Collapse
|
4
|
Šutković J. Neutrophils and COVID-19. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:347-384. [PMID: 40246349 DOI: 10.1016/bs.pmbts.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Neutrophils are the first line of defense against pathogens, most effectively by forming Neutrophil Extracellular Traps (NETs). Neutrophiles are further classified into several subpopulations during their development, eliminating pathogens through various mechanisms. However, due to the chaotic and uncontrolled immune response, NETs are often severely resulting in tissue damage and lung infections. The uncontrolled and poorly acknowledged host response regarding the cytokine storm is one of the major causes of severe COVID-19 conditions. Specifically, the increased formation of low-density neutrophils (LDNs), together with neutrophil extracellular traps (NETs) is closely linked with the severity and poor prognosis in patients with COVID-19. In this review, we discuss in detail the ontogeny of neutrophils at different stages and their recruitment and activation after infections, focusing on SARS-CoV-2. In addition, this chapter summarized the research progress on potential targeted drugs (NETs and Cytokine inhibitors) for neutrophil medical therapy and hoped to provide reference for the development of related therapeutic drugs for critically ill COVID-19 patients.
Collapse
Affiliation(s)
- Jasmin Šutković
- Department Genetics and Bioegnineering, International University of Sarajevo, Hrasnička cesta, Bosnia & Herzegovina.
| |
Collapse
|
5
|
Zakrzewski P, Rice CM, Fleming K, Cela D, Groves SJ, Ponce-Garcia FM, Gibbs W, Roberts K, Pike T, Strathdee D, Anderson E, Nobbs AH, Toye AM, Steward C, Amulic B. Tafazzin regulates neutrophil maturation and inflammatory response. EMBO Rep 2025; 26:1590-1619. [PMID: 39962231 PMCID: PMC11933368 DOI: 10.1038/s44319-025-00393-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 03/26/2025] Open
Abstract
Barth syndrome (BTHS) is a rare genetic disease caused by mutations in the TAFAZZIN gene. It is characterized by neutropenia, cardiomyopathy and skeletal myopathy. Neutropenia in BTHS is associated with life-threatening infections, yet there is little understanding of the molecular and physiological causes of this phenomenon. We combined bone marrow analysis, CRISPR/Cas9 genome editing in hematopoietic stem cells and functional characterization of circulating BTHS patient neutrophils to investigate the role of TAFAZZIN in neutrophils and their progenitors. We demonstrate a partial cell intrinsic differentiation defect, along with a dysregulated neutrophil inflammatory response in BTHS, including elevated degranulation and formation of neutrophil extracellular traps (NETs) in response to calcium flux. Developmental and functional alterations in BTHS neutrophils are underpinned by perturbations in the unfolded protein response (UPR) signaling pathway, suggesting potential therapeutic avenues for targeting BTHS neutropenia.
Collapse
Affiliation(s)
- Przemysław Zakrzewski
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Christopher M Rice
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Kathryn Fleming
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Drinalda Cela
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Sarah J Groves
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Fernando M Ponce-Garcia
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Willem Gibbs
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Kiran Roberts
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Tobias Pike
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Eve Anderson
- Cancer Research UK Scotland Institute, Glasgow, G61 1BD, UK
| | - Angela H Nobbs
- Bristol Dental School Research Laboratories, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, UK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Colin Steward
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK
| | - Borko Amulic
- School of Cellular and Molecular Medicine, Biomedical Sciences Building, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
6
|
Manetsch P, Hottiger MO. Unleashing viral mimicry: A combinatorial strategy to enhance the efficacy of PARP7 inhibitors. Bioessays 2025; 47:e2400087. [PMID: 39502005 PMCID: PMC11755700 DOI: 10.1002/bies.202400087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/03/2024] [Accepted: 10/23/2024] [Indexed: 01/24/2025]
Abstract
Cancer cells exploit mechanisms to evade immune detection triggered by aberrant self-nucleic acids (NA). PARP7, a key player in this immune evasion strategy, has emerged as a potential target for cancer therapy. PARP7 inhibitors reactivate NA sensing, resulting in type I interferon (IFN) signaling, programmed cell death, anti-tumor immunity, and tumor regression. Cancer cells with elevated IFN-stimulated gene (ISG) scores, representing a viral mimicry-primed state, are particularly sensitive to PARP7 inhibition. This review focuses on the endogenous sources of NA in cancer and the potential to exploit elevated aberrant self-NA in cancer therapy. We describe strategies to increase cytoplamic NA levels, including targeting epigenetic control, DNA damage response, and mitochondrial function. We also discuss targeting RNA processing pathways, such as splicing and RNA editing, to enhance the immunostimulatory potential of existing NA. Combining PARP7 inhibitors with NA elevating strategies may improve cancer immunotherapy, especially for tumors with high ISG scores.
Collapse
Affiliation(s)
- Patrick Manetsch
- Department of Molecular Mechanisms of DiseaseUniversity of ZurichZurichSwitzerland
- Molecular Life Science PhD Program of the Life Science Zurich Graduate SchoolUniversity of ZurichZurichSwitzerland
| | - Michael O. Hottiger
- Department of Molecular Mechanisms of DiseaseUniversity of ZurichZurichSwitzerland
| |
Collapse
|
7
|
Xia L, Yan X, Zhang H. Mitochondrial DNA-activated cGAS-STING pathway in cancer: Mechanisms and therapeutic implications. Biochim Biophys Acta Rev Cancer 2025; 1880:189249. [PMID: 39701325 DOI: 10.1016/j.bbcan.2024.189249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024]
Abstract
Mitochondrial DNA (mtDNA), a circular double-stranded DNA located within mitochondria, plays a pivotal role in mitochondrial-induced innate immunity, particularly via the cyclic GMP-AMP synthase (cGAS)-STING pathway, which recognizes double-stranded DNA and is crucial for pathogen resistance. Recent studies elucidate the interplay among mtDNA, the cGAS-STING pathway, and neutrophil extracellular traps (NETs) in the context of cancer. mtDNA uptake by recipient cells activates the cGAS-STING pathway, while mtDNA leakage reciprocally regulates NET release, amplifying inflammation and promoting NETosis, a mechanism of tumor cell death. Autophagy modulates these processes by clearing damaged mitochondria and degrading cGAS, thus preventing mtDNA recognition. Tumor microenvironmental factors, such as metabolic reprogramming and lipid accumulation, induce mitochondrial stress, ROS production, and further mtDNA leakage. This review explores strategies in cancer drug development that leverage mtDNA leakage to activate the cGAS-STING pathway, potentially converting 'cold tumors' into 'hot tumors,' while discussing advancements in targeted therapies and proposing new research methodologies.
Collapse
Affiliation(s)
- Lintao Xia
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiuli Yan
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Hui Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
8
|
Quan M, Zhang H, Deng X, Liu H, Xu Y, Song X. Neutrophils, NETs and multiple sclerosis: a mini review. Front Immunol 2025; 16:1487814. [PMID: 39935468 PMCID: PMC11810747 DOI: 10.3389/fimmu.2025.1487814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025] Open
Abstract
Multiple sclerosis (MS), a chronic inflammatory and degenerative autoimmune disease characterized by the activation of various inflammatory cells, leads to demyelination and neuronal injury. Neutrophils, often underestimated in MS, are gaining increased attention for their significant functions in MS patients and the experimental autoimmune encephalomyelitis (EAE) animal model. Neutrophils play multiple roles in mediating the pathogenesis of autoimmune diseases, and numerous studies suggest that neutrophils might have a crucial role through neutrophil extracellular trap (NET) formation. Studies on NETs in MS are still in their infancy. In this review, we discuss the clinical perspective on the linkage between neutrophils and MS or EAE, as well as the role of NETs in the pathogenesis of MS/EAE. Further, we analyze the potential mechanisms by which NETs contribute to MS, the protective effects of NETs in MS, and their value as targets for disease intervention. NET formation and/or clearance as a therapeutic approach for MS still requires research in greater depth.
Collapse
Affiliation(s)
- Moyuan Quan
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| | - Huining Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| | - Xiaohong Deng
- Department of Rehabilitation Medicine, Beijing Zhongguancun Hospital, Beijing, China
| | - Huijia Liu
- Department of Internal Medicine, The Military Special Care Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Yanqiu Xu
- Department of Neurology, The Third Hospital of Shijiazhuang, Shijiazhuang, Hebei, China
| | - Xiujuan Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Key Laboratory of Neurology of Hebei Province, Shijiazhuang, Hebei, China
| |
Collapse
|
9
|
Sun T, Wang P, Zhai X, Wang Z, Miao X, Yang Y, Wu J. Neutrophil extracellular traps induce barrier dysfunction in DSS-induced ulcerative colitis via the cGAS-STING pathway. Int Immunopharmacol 2024; 143:113358. [PMID: 39388893 DOI: 10.1016/j.intimp.2024.113358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/20/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
Peptidyl arginine deiminase 4 (PAD4)-mediated neutrophil extracellular traps (NETs) play a crucial role in the pathogenesis of ulcerative colitis (UC). The cGAS-STING intracellular DNA-sensing pathway has been recognized as a pivotal mediator of inflammation. This study aimed to explore how NETs contribute to intestinal inflammation and barrier dysfunction in UC, focusing on the cGAS-STING pathway. We observed a significant increase of STING expression in UC mouse colons, which was mitigated by blocking NET formation through PAD4 genetic knockout. Moreover, NETs were discovered to activate the cGAS-STING pathway in MC38 cells in a dose and time-dependent manner, leading to the secretion of inflammatory cytokines and impaired barrier function. Additionally, STING deficiency ameliorated the clinical colitis index, intestinal inflammation, and barrier dysfunction. These findings underscore the involvement of cGAS-STING in regulating NET-mediated intestinal inflammation, suggesting its potential as a novel therapeutic target for UC.
Collapse
Affiliation(s)
- Tao Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ping Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinru Zhai
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhiwei Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinyu Miao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yang Yang
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China.
| | - Jie Wu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
10
|
Cyr P, Fader LD, Burch JD, Pike KA, Sietsema DV, Boily MO, Ciblat S, Sgarioto N, Skeldon AM, Gaudreault S, Le Gros P, Dumais V, McKay DJJ, Abraham NS, Seliniotakis R, Beveridge RE. Discovery of Potent and Orally Bioavailable Pyrimidine Amide cGAS Inhibitors via Structure-Guided Hybridization. ACS Med Chem Lett 2024; 15:2201-2209. [PMID: 39691514 PMCID: PMC11647726 DOI: 10.1021/acsmedchemlett.4c00471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024] Open
Abstract
Using a high-throughput screening (HTS) approach, a new GTP-site binding pyridine-carboxylate series of cGAS inhibitors was discovered. The biochemical potency of this new pyridine carboxylate series was improved 166-fold from the original hit to double-digit nanomolar levels using structure-based design insights, but the series was found to suffer from low permeability and low bioavailability. A structure-based hybridization of the metal-binding motifs of the pyridine carboxylate series and our previously disclosed tetrahydrocarboline GTP-site ligand 23 identified pyrimidine amide compound 36. Compound 36 is potent against both human and mouse cGAS isoforms and has a favorable pharmacokinetic (PK) profile in mice. Additionally, compound 36 displayed a dose-dependent reduction in cGAMP production in a ConA pharmacodynamic mouse model of acute liver injury, demonstrating potential utility as an in vivo tool compound for further investigation of the cGAS pathway.
Collapse
Affiliation(s)
- Patrick Cyr
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Lee D. Fader
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Jason D. Burch
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Kelly A. Pike
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Daniel V. Sietsema
- Ventus
Therapeutics, 100 Beaver
St, Suite 201, Waltham, Massachusetts 02453, United States
| | - Marc-Olivier Boily
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Stéphane Ciblat
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Nicolas Sgarioto
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | | | - Samuel Gaudreault
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Philippe Le Gros
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Valérie Dumais
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Daniel J. J. McKay
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Nathan S. Abraham
- Ventus
Therapeutics, 100 Beaver
St, Suite 201, Waltham, Massachusetts 02453, United States
| | - Ria Seliniotakis
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| | - Ramsay E. Beveridge
- Ventus
Therapeutics, 4800 rue Lévy #110, Saint-Laurent H4R 2P1, Quebec, Canada
| |
Collapse
|
11
|
Lu CH, Shen CY, Li KJ, Wu CH, Chen YH, Kuo YM, Hsieh SC, Yu CL. Resolution of acute inflammation induced by monosodium urate crystals (MSU) through neutrophil extracellular trap-MSU aggregate-mediated negative signaling. J Inflamm (Lond) 2024; 21:50. [PMID: 39605016 PMCID: PMC11604016 DOI: 10.1186/s12950-024-00423-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Polymorphonuclear neutrophils (PMN) activation by monosodium urate crystals (MSU) is crucial to acute gouty arthritis and subsequent spontaneous remission within 7-10 days. Activated PMNs release neutrophil extracellular traps (NETs) that entrap MSU crystals, forming NET-MSU aggregates. Whether NET-MSU aggregates contribute to the resolution of acute inflammation remains to be elucidated. This study uses a cell-based approach to unveil their molecular bases. METHODS All-trans retinoic acid-differentiated HL-60 cells (dHL-60) served as surrogate PMNs. NET release from MSU-activated dHL-60 was confirmed by detecting DNA, neutrophil elastase, and citrullinated histone 3, forming large NET-MSU aggregates. NET area was measured with Fiji software after SYTOX Green staining. Released pro-inflammatory cytokines IL-8 and TNF-α, and the anti-inflammatory cytokine IL-1RA in culture supernatants were quantified to calculate the estimate inflammation score (EIS). Cellular redox state was determined by a FRET-based sensor. Expression of intracellular positive (ERK1/2) and negative (SHP-1 and SHIP-1) cytokine signaling regulators was detected by western blot. qPCR detected mRNA expressions of CISH and SOCS1-SOCS7. Flow cytometry measured neutrophil N1 (CD54) and N2 (CD182) surface markers after staining with fluorescent-conjugated antibodies. RESULTS Incubating dHL-60 with MSU for 4 h maximized NET-MSU aggregate formation and acute inflammation with an EIS of 11.6. Prolonging the incubation of dHL-60 + MSU to 22 h gradually raised the EIS to 19.40 without increasing NET area, due to reduced cellular redox capacity. Adding both new dHL-60 and new MSU crystals to the culture, mimicking the clinical scenario, increased NET area but conversely suppressed EIS to 1.53, indicating acute inflammation resolution. The resolution of acute inflammation following prolonged incubation was attributed to decreases in P-ERK and increases in P-SHP-1, SOCS2, SOCS3, and CISH gene expressions, which may suppress pro-inflammatory and enhance anti-inflammatory cytokine production. Moreover, the large NET-MSU aggregates facilitated N1 to N2 polarization, crucial for accelerating inflammation resolution. CONCLUSION We explored the potential molecular basis for the spontaneous resolution of MSU induced acute inflammation using a cell-based model in that huge NET-MSU aggregates frustrate the transformation of newly entering PMNs to the N2 phenotype, enhancing the production of the anti-inflammatory cytokine IL-1RA.
Collapse
Affiliation(s)
- Cheng-Hsun Lu
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10002, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, No.1, Chang-Te Street, Taipei, 10048, Taiwan
| | - Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10002, Taiwan
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10002, Taiwan
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital-Hsinchu Biomedical Park, No. 2, Sec. 1, Shengyi Road., Zhubei City, Hsinchu County, 302058, Taiwan
| | - Yu-Hsuan Chen
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10002, Taiwan
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10002, Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, No.1, Chang-Te Street, Taipei, 10048, Taiwan
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10002, Taiwan
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 10002, Taiwan.
| |
Collapse
|
12
|
Huang Y, Jiang W, Zhou R. DAMP sensing and sterile inflammation: intracellular, intercellular and inter-organ pathways. Nat Rev Immunol 2024; 24:703-719. [PMID: 38684933 DOI: 10.1038/s41577-024-01027-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 05/02/2024]
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous molecules that are released from host cells as a result of cell death or damage. The release of DAMPs in tissues is associated with loss of tissue homeostasis. Sensing of DAMPs by innate immune receptors triggers inflammation, which can be beneficial in initiating the processes that restore tissue homeostasis but can also drive inflammatory diseases. In recent years, the sensing of intracellular DAMPs has received extensive attention in the field of sterile inflammation. However, emerging studies have shown that DAMPs that originate from neighbouring cells, and even from distal tissues or organs, also mediate sterile inflammatory responses. This multi-level sensing of DAMPs is crucial for intercellular, trans-tissue and trans-organ communication. Here, we summarize how DAMP-sensing receptors detect DAMPs from intracellular, intercellular or distal tissue and organ sources to mediate sterile inflammation. We also discuss the possibility of targeting DAMPs or their corresponding receptors to treat inflammatory diseases.
Collapse
Affiliation(s)
- Yi Huang
- Key Laboratory of Immune Response and Immunotherapy, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Wei Jiang
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
- Key Laboratory of Immune Response and Immunotherapy, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
13
|
Mihlan M, Wissmann S, Gavrilov A, Kaltenbach L, Britz M, Franke K, Hummel B, Imle A, Suzuki R, Stecher M, Glaser KM, Lorentz A, Carmeliet P, Yokomizo T, Hilgendorf I, Sawarkar R, Diz-Muñoz A, Buescher JM, Mittler G, Maurer M, Krause K, Babina M, Erpenbeck L, Frank M, Rambold AS, Lämmermann T. Neutrophil trapping and nexocytosis, mast cell-mediated processes for inflammatory signal relay. Cell 2024; 187:5316-5335.e28. [PMID: 39096902 DOI: 10.1016/j.cell.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 04/10/2024] [Accepted: 07/08/2024] [Indexed: 08/05/2024]
Abstract
Neutrophils are sentinel immune cells with essential roles for antimicrobial defense. Most of our knowledge on neutrophil tissue navigation derived from wounding and infection models, whereas allergic conditions remained largely neglected. Here, we analyzed allergen-challenged mouse tissues and discovered that degranulating mast cells (MCs) trap living neutrophils inside them. MCs release the attractant leukotriene B4 to re-route neutrophils toward them, thus exploiting a chemotactic system that neutrophils normally use for intercellular communication. After MC intracellular trap (MIT) formation, neutrophils die, but their undigested material remains inside MC vacuoles over days. MCs benefit from MIT formation, increasing their functional and metabolic fitness. Additionally, they are more pro-inflammatory and can exocytose active neutrophilic compounds with a time delay (nexocytosis), eliciting a type 1 interferon response in surrounding macrophages. Together, our study highlights neutrophil trapping and nexocytosis as MC-mediated processes, which may relay neutrophilic features over the course of chronic allergic inflammation.
Collapse
Affiliation(s)
- Michael Mihlan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany.
| | - Stefanie Wissmann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute for Biomechanics, ETH Zürich, Zürich 8092, Switzerland
| | - Alina Gavrilov
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Roche Pharma Research and Early Development (pRED), Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology (CMI2O), Roche Innovation Center, Basel 4070, Switzerland
| | - Lukas Kaltenbach
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Marie Britz
- Department of Dermatology, Universitätsklinikum Münster, Münster 48149, Germany
| | - Kristin Franke
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Barbara Hummel
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Andrea Imle
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg 69117, Germany
| | - Ryo Suzuki
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | - Manuel Stecher
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany
| | - Katharina M Glaser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institut Curie, PSL Research University, INSERM U932, Paris 75005, France
| | - Axel Lorentz
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart 70593, Germany
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Center for Biotechnology, Khalifa University of Science and Technology, PO Box 127788, Abu Dhabi, United Arab Emirates
| | - Takehiko Yokomizo
- Department of Biochemistry, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center and Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Ritwick Sawarkar
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Medical Research Council (MRC) Toxicology Unit and Department of Genetics, University of Cambridge, Cambridge CB21QR, UK
| | - Alba Diz-Muñoz
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory (EMBL), Heidelberg 69117, Germany
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Gerhard Mittler
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Marcus Maurer
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Karoline Krause
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Magda Babina
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Berlin 12203, Germany; Charité-Universitätsmedizin Berlin, Institute of Allergology, Berlin 12203, Germany
| | - Luise Erpenbeck
- Department of Dermatology, Universitätsklinikum Münster, Münster 48149, Germany
| | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Rostock 18057, Germany; Department Life, Light and Matter, Rostock University, Rostock 18051, Germany
| | - Angelika S Rambold
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg 79108, Germany; Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Münster 48149, Germany.
| |
Collapse
|
14
|
Brockman ML, Scruggs TA, Wang L, Kabboul G, Calvert JW, Levit RD. The Cardioprotective Role of Neutrophil-Specific STING in Myocardial Ischemia/Reperfusion Injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611551. [PMID: 39314411 PMCID: PMC11418936 DOI: 10.1101/2024.09.06.611551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Background Neutrophils are the most rapid and abundant immune cells to infiltrate the myocardium following myocardial ischemia/reperfusion injury (MI/R). Neutrophil heterogeneity has not been well characterized in MI/R, and studies have shown conflicting results regarding the impact of neutrophil depletion on cardiac injury. We thus aim to study the impact of neutrophils with enriched type I interferon signature and the role of STING (stimulator of interferon genes) signaling in neutrophils on cardiac reperfusion injury. Methods We utilized single-cell RNA sequencing to study neutrophil heterogeneity in response to MI/R. We generated a neutrophil-specific STING knockout mouse to assess the role of neutrophil STING in a model of MI/R. We examined cardiac function following injury via echocardiography and assessed the immune cell trajectory following injury utilizing flow cytometry. Results We identified a population of neutrophils with enriched type I interferon signaling and response to type I interferon following MI/R. We found that genetic deletion of neutrophil-specific STING led to worsened cardiac function following MI/R. Further investigation of the immune response by flow cytometry revealed decreased neutrophil infiltration into the myocardium and a shift in macrophage polarization. Conclusions Our findings suggest that neutrophil-specific STING is cardioprotective in MI/R, partly due to its effects on downstream immune cells. These results demonstrate that early alterations or therapeutic interventions can influence key events in the resolution of inflammation following MI/R.
Collapse
Affiliation(s)
- Maegan L. Brockman
- Department of Medicine; Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Triniti A. Scruggs
- Department of Medicine; Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lanfang Wang
- Department of Medicine; Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Gabriella Kabboul
- Department of Medicine; Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University
| | - John W. Calvert
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Rebecca D. Levit
- Department of Medicine; Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
15
|
Murata K, Murao A, Tan C, Wang P, Aziz M. B-1a cells scavenge NETs to attenuate sepsis. J Leukoc Biol 2024; 116:632-643. [PMID: 38484156 PMCID: PMC11367732 DOI: 10.1093/jleuko/qiae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 09/03/2024] Open
Abstract
B-1a cells, a regulatory subset of B lymphocytes, produce natural IgM and interleukin-10. Neutrophil extracellular traps (NETs) play a crucial role in pathogen defense, but their excessive formation during sepsis can cause further inflammation and tissue damage. In sepsis, extracellular cold-inducible RNA-binding protein (eCIRP), a damage-associated molecular pattern, is released to induce NET formation. We hypothesize that B-1a cells clear NETs to prevent sepsis-induced injury. Sepsis in mice was induced by injecting 1 × 107 and 5 × 107 colony-forming units of Escherichia coli intraperitoneally. After 4 and 20 h, we assessed the number of B-1a cells in the peritoneal cavity using flow cytometry. Our results showed that the number of peritoneal B-1a cells was significantly decreased in E. coli sepsis mice. Importantly, replenishing B-1a cells via intraperitoneal injection in sepsis mice significantly decreased NETs in peritoneal neutrophils. We also observed a decrease in serum inflammation and injury markers and a significant increase in the overall survival rate in B-1a cell-treated septic mice. To understand the mechanism, we cocultured bone marrow-derived neutrophils with peritoneal B-1a cells in a contact or noncontact condition using an insert and stimulated them with eCIRP. After 4 h, we found that eCIRP significantly increased NET formation in bone marrow-derived neutrophils. Interestingly, we observed that B-1a cells inhibited NETs by 67% in a contact-dependent manner. Surprisingly, when B-1a cells were cultured in inserts, there was no significant decrease in NET formation, suggesting that direct cell-to-cell contact is crucial for this inhibitory effect. We further determined that B-1a cells promoted NET phagocytosis, and this was mediated through natural IgM, as blocking the IgM receptor attenuated the engulfment of NETs by B-1a cells. Finally, we identified that following their engulfment, NETs were localized into the lysosomal compartment for lysis. Thus, our study suggests that B-1a cells decrease NET content in eCIRP-treated neutrophils and E. coli sepsis mice.
Collapse
Affiliation(s)
- Kensuke Murata
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Atsushi Murao
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Chuyi Tan
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
| | - Ping Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, 350 Community Drive, Manhasset, NY 11030, United States
| |
Collapse
|
16
|
Malamud M, Whitehead L, McIntosh A, Colella F, Roelofs AJ, Kusakabe T, Dambuza IM, Phillips-Brookes A, Salazar F, Perez F, Shoesmith R, Zakrzewski P, Sey EA, Rodrigues C, Morvay PL, Redelinghuys P, Bedekovic T, Fernandes MJG, Almizraq R, Branch DR, Amulic B, Harvey J, Stewart D, Yuecel R, Reid DM, McConnachie A, Pickering MC, Botto M, Iliev ID, McInnes IB, De Bari C, Willment JA, Brown GD. Recognition and control of neutrophil extracellular trap formation by MICL. Nature 2024; 633:442-450. [PMID: 39143217 PMCID: PMC11390483 DOI: 10.1038/s41586-024-07820-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 07/11/2024] [Indexed: 08/16/2024]
Abstract
Regulation of neutrophil activation is critical for disease control. Neutrophil extracellular traps (NETs), which are web-like structures composed of DNA and neutrophil-derived proteins, are formed following pro-inflammatory signals; however, if this process is uncontrolled, NETs contribute to disease pathogenesis, exacerbating inflammation and host tissue damage1,2. Here we show that myeloid inhibitory C-type lectin-like (MICL), an inhibitory C-type lectin receptor, directly recognizes DNA in NETs; this interaction is vital to regulate neutrophil activation. Loss or inhibition of MICL functionality leads to uncontrolled NET formation through the ROS-PAD4 pathway and the development of an auto-inflammatory feedback loop. We show that in the context of rheumatoid arthritis, such dysregulation leads to exacerbated pathology in both mouse models and in human patients, where autoantibodies to MICL inhibit key functions of this receptor. Of note, we also detect similarly inhibitory anti-MICL autoantibodies in patients with other diseases linked to aberrant NET formation, including lupus and severe COVID-19. By contrast, dysregulation of NET release is protective during systemic infection with the fungal pathogen Aspergillus fumigatus. Together, we show that the recognition of NETs by MICL represents a fundamental autoregulatory pathway that controls neutrophil activity and NET formation.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/metabolism
- Aspergillus fumigatus/immunology
- Aspergillus fumigatus/pathogenicity
- Autoantibodies/immunology
- Autoantibodies/pharmacology
- COVID-19/immunology
- COVID-19/virology
- Disease Models, Animal
- DNA/metabolism
- DNA/immunology
- Extracellular Traps/metabolism
- Extracellular Traps/immunology
- Feedback, Physiological
- Inflammation/immunology
- Inflammation/metabolism
- Lectins, C-Type/antagonists & inhibitors
- Lectins, C-Type/deficiency
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/metabolism
- Mice, Inbred C57BL
- Neutrophil Activation
- Neutrophils/immunology
- Neutrophils/metabolism
- Protein-Arginine Deiminase Type 4/metabolism
- Reactive Oxygen Species/metabolism
- Receptors, Mitogen/antagonists & inhibitors
- Receptors, Mitogen/deficiency
- Receptors, Mitogen/immunology
- Receptors, Mitogen/metabolism
Collapse
Affiliation(s)
- Mariano Malamud
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Lauren Whitehead
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Alasdair McIntosh
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Fabio Colella
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Anke J Roelofs
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Takato Kusakabe
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York City, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease (JRI), Weill Cornell Medicine, New York City, NY, USA
| | - Ivy M Dambuza
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Fabián Salazar
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Federico Perez
- Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Romey Shoesmith
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Emily A Sey
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Petruta L Morvay
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | - Tina Bedekovic
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Maria J G Fernandes
- Faculty of Medicine, Department of Microbiology, Infectious Diseases, and Immunology, Laval University, Quebec City, Quebec, Canada
| | - Ruqayyah Almizraq
- Medical Affairs and Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Donald R Branch
- Medical Affairs and Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Borko Amulic
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, UK
| | - Jamie Harvey
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Diane Stewart
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Raif Yuecel
- Centre for Cytomics, University of Exeter, Exeter, UK
| | - Delyth M Reid
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Alex McConnachie
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Matthew C Pickering
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Marina Botto
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Iliyan D Iliev
- Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York City, NY, USA
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease (JRI), Weill Cornell Medicine, New York City, NY, USA
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Cosimo De Bari
- Centre for Arthritis and Musculoskeletal Health, University of Aberdeen, Aberdeen, UK
| | - Janet A Willment
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Gordon D Brown
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK.
- Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
17
|
Wei C, Huang Q, Zeng F, Ma L, Bai X, Zhu X, Gao H, Qi X. Cyclic guanosine monophosphate-adenosine monophosphate synthetase/stimulator of interferon genes signaling aggravated corneal allograft rejection through neutrophil extracellular traps. Am J Transplant 2024; 24:1583-1596. [PMID: 38648890 DOI: 10.1016/j.ajt.2024.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
The activation of innate immunity following transplantation has been identified as a crucial factor in allograft inflammation and rejection. However, the role of cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)/stimulator of interferon genes (STING) signaling-mediated innate immunity in the pathogenesis of allograft rejection remains unclear. Utilizing a well-established murine model of corneal transplantation, we demonstrated increased expression of cGAS and STING in rejected-corneal allografts compared with syngeneic (Syn) and normal (Nor) corneas, along with significant activation of the cGAS/STING pathway, as evidenced by the enhanced phosphorylation of TANK-binding kinase 1and interferon regulatory factor 3. Pharmacological and genetic inhibition of cGAS/STING signaling markedly delayed corneal transplantation rejection, resulting in prolonged survival time and reduced inflammatory infiltration. Furthermore, we observed an increase in the formation of neutrophil extracellular traps (NETs) in rejected allografts, and the inhibition of NET formation through targeting peptidylarginine deiminase 4 and DNase I treatment significantly alleviated immune rejection and reduced cGAS/STING signaling activity. Conversely, subconjunctival injection of NETs accelerated corneal transplantation rejection and enhanced the activation of the cGAS/STING pathway. Collectively, these findings demonstrate that NETs contribute to the exacerbation of allograft rejection via cGAS/STING signaling, highlighting the targeting of the NETs/cGAS/STING signaling pathway as a potential strategy for prolonging allograft survival.
Collapse
Affiliation(s)
- Chao Wei
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Shandong First Medical University, Qingdao, Shandong, China
| | - Qing Huang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Shandong First Medical University, Qingdao, Shandong, China
| | - Fanxing Zeng
- Refractive Surgery Center, Guangzhou Huangpu Aier Eye Hospital, Guangzhou, Guangdong, China
| | - Li Ma
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Shandong First Medical University, Qingdao, Shandong, China
| | - Xiaofei Bai
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Shandong First Medical University, Qingdao, Shandong, China
| | - Xuejing Zhu
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Medical Department of Qingdao University, Qingdao, Shandong, China
| | - Hua Gao
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Eye Institute of Shandong First Medical University, School of Ophthalmology, Shandong First Medical University, Jinan, Shandong, China
| | - Xiaolin Qi
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Medical Department of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
18
|
Chang L. Harnessing cGAS-STING axis for therapeutic benefits in systemic lupus erythematosus. Int J Rheum Dis 2024; 27:e15256. [PMID: 38982864 DOI: 10.1111/1756-185x.15256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 07/11/2024]
Abstract
The cyclic GMP-AMP synthase (cGAS), a prominent intracellular DNA sensor in mammalian cells, controls the innate immune response and the stimulator of interferon genes (STING)-mediated synthesis of pro-inflammatory cytokines, such as type-I interferon (IFN-I). For decades, IFN-I has been hypothesized to be essential in the development of systemic lupus erythematosus (SLE), a chronic multisystem autoimmunity characterized by immune complex (IC) deposition in small vessels. Recent findings revealed that the activation of the cGAS-STING pathway by self-DNA would propagate the autoimmune responses via upregulating IFN-I production in SLE. In this review, we aimed to provide a comprehensive outlook of the role of the cGAS-STING pathway in SLE pathobiology, as well as, a better understanding of current therapeutic opportunities targeting this axis.
Collapse
Affiliation(s)
- Liu Chang
- Department of Rheumatology, Henan Provincial Hospital of Traditional Chinese Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Nofi CP, Prince JM, Wang P, Aziz M. Chromatin as alarmins in necrotizing enterocolitis. Front Immunol 2024; 15:1403018. [PMID: 38881893 PMCID: PMC11176418 DOI: 10.3389/fimmu.2024.1403018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/20/2024] [Indexed: 06/18/2024] Open
Abstract
Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease primarily affecting premature neonates, marked by poorly understood pro-inflammatory signaling cascades. Recent advancements have shed light on a subset of endogenous molecular patterns, termed chromatin-associated molecular patterns (CAMPs), which belong to the broader category of damage-associated molecular patterns (DAMPs). CAMPs play a crucial role in recognizing pattern recognition receptors and orchestrating inflammatory responses. This review focuses into the realm of CAMPs, highlighting key players such as extracellular cold-inducible RNA-binding protein (eCIRP), high mobility group box 1 (HMGB1), cell-free DNA, neutrophil extracellular traps (NETs), histones, and extracellular RNA. These intrinsic molecules, often perceived as foreign, have the potential to trigger immune signaling pathways, thus contributing to NEC pathogenesis. In this review, we unravel the current understanding of the involvement of CAMPs in both preclinical and clinical NEC scenarios. We also focus on elucidating the downstream signaling pathways activated by these molecular patterns, providing insights into the mechanisms that drive inflammation in NEC. Moreover, we scrutinize the landscape of targeted therapeutic approaches, aiming to mitigate the impact of tissue damage in NEC. This in-depth exploration offers a comprehensive overview of the role of CAMPs in NEC, bridging the gap between preclinical and clinical insights.
Collapse
Affiliation(s)
- Colleen P. Nofi
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Jose M. Prince
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
- Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
- Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
20
|
Yang Y, Qi J, Hu J, Zhou Y, Zheng J, Deng W, Inam M, Guo J, Xie Y, Li Y, Xu C, Deng W, Chen W. Lovastatin/SN38 co-loaded liposomes amplified ICB therapeutic effect via remodeling the immunologically-cold colon tumor and synergized stimulation of cGAS-STING pathway. Cancer Lett 2024; 588:216765. [PMID: 38408604 DOI: 10.1016/j.canlet.2024.216765] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/17/2024] [Accepted: 02/22/2024] [Indexed: 02/28/2024]
Abstract
Current immune checkpoint blockade (ICB) immunotherapeutics have revolutionized cancer treatment. However, many cancers especially the "immunologically cold" tumors, do not respond to ICB, prompting the search for additional strategies to achieve durable responses. The cGAS-STING pathway, as an essential immune response pathway, has been demonstrated for a potent target to sensitize ICB immunotherapy. However, the low efficiency of conventional STING agonists limits their clinical application. Recent studies have shown that DNA topoisomerase I (TOPI) inhibitor chemodrug SN38 can activate the cGAS-STING pathway and induce an immune response through DNA damage, while the traditional statins medication lovastatin was found to inhibit DNA damage repair, which may in turn upregulate the damaged DNA level. Herein, we have developed a liposomal carrier co-loaded with SN38 and lovastatin (SL@Lip), which can be accumulated in tumors and efficiently released SN38 and lovastatin, addressing the problem of weak solubility of these two drugs. Importantly, lovastatin can increase DNA damage and enhance the activation of cGAS-STING pathway, coordinating with SN38 chemotherapy and exhibiting the enhanced combinational immunotherapy of PD-1 antibody by remodeling the tumor microenvironment in mouse colorectal cancer of both subcutaneous and orthotopic xenograft models. Overall, this study demonstrates that lovastatin-assisted cGAS-STING stimulation mediated by liposomal delivery system significantly strengthened both chemotherapy and immunotherapy of colorectal cancer, providing a clinically translational strategy for combinational ICB therapy in the "immunologically cold" tumors.
Collapse
Affiliation(s)
- Yi Yang
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jialong Qi
- Yunnan Digestive Endoscopy Clinical Medical Center, Department of Gastroenterology, The First People's Hospital of Yunnan Province, Kunming, 650032, PR China
| | - Jialin Hu
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - You Zhou
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jiena Zheng
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Wenxia Deng
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Muhammad Inam
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Jiaxin Guo
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yongyi Xie
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Yuan Li
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China
| | - Chuanshan Xu
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| | - Wenjie Chen
- School of Pharmaceutical Science, State Key Laboratory of Respiratory Disease & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR China.
| |
Collapse
|
21
|
Li B, Xu L, Wang Z, Shi Q, Cui Y, Fan W, Wu Q, Tong X, Yan H. Neutrophil Extracellular Traps Regulate Surgical Brain Injury by Activating the cGAS-STING Pathway. Cell Mol Neurobiol 2024; 44:36. [PMID: 38637346 PMCID: PMC11026279 DOI: 10.1007/s10571-024-01470-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/15/2024] [Indexed: 04/20/2024]
Abstract
Surgical brain injury (SBI), induced by neurosurgical procedures or instruments, has not attracted adequate attention. The pathophysiological process of SBI remains sparse compared to that of other central nervous system diseases thus far. Therefore, novel and effective therapies for SBI are urgently needed. In this study, we found that neutrophil extracellular traps (NETs) were present in the circulation and brain tissues of rats after SBI, which promoted neuroinflammation, cerebral edema, neuronal cell death, and aggravated neurological dysfunction. Inhibition of NETs formation by peptidylarginine deiminase (PAD) inhibitor or disruption of NETs with deoxyribonuclease I (DNase I) attenuated SBI-induced damages and improved the recovery of neurological function. We show that SBI triggered the activation of cyclic guanosine monophosphate-adenosine monophosphate synthase stimulator of interferon genes (cGAS-STING), and that inhibition of the cGAS-STING pathway could be beneficial. It is worth noting that DNase I markedly suppressed the activation of cGAS-STING, which was reversed by the cGAS product cyclic guanosine monophosphate-adenosine monophosphate (cGMP-AMP, cGAMP). Furthermore, the neuroprotective effect of DNase I in SBI was also abolished by cGAMP. NETs may participate in the pathophysiological regulation of SBI by acting through the cGAS-STING pathway. We also found that high-dose vitamin C administration could effectively inhibit the formation of NETs post-SBI. Thus, targeting NETs may provide a novel therapeutic strategy for SBI treatment, and high-dose vitamin C intervention may be a promising translational therapy with an excellent safety profile and low cost.
Collapse
Affiliation(s)
- Bingbing Li
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Lixia Xu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Zhengang Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Qi Shi
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Yang Cui
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Weijia Fan
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Qiaoli Wu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China
| | - Xiaoguang Tong
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| | - Hua Yan
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin, 300350, China.
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300350, China.
| |
Collapse
|
22
|
Wang C, Kaur K, Xu C, Abu-Amer Y, Mbalaviele G. Chemotherapy activates inflammasomes to cause inflammation-associated bone loss. eLife 2024; 13:RP92885. [PMID: 38602733 PMCID: PMC11008812 DOI: 10.7554/elife.92885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024] Open
Abstract
Chemotherapy is a widely used treatment for a variety of solid and hematological malignancies. Despite its success in improving the survival rate of cancer patients, chemotherapy causes significant toxicity to multiple organs, including the skeleton, but the underlying mechanisms have yet to be elucidated. Using tumor-free mouse models, which are commonly used to assess direct off-target effects of anti-neoplastic therapies, we found that doxorubicin caused massive bone loss in wild-type mice, a phenotype associated with increased number of osteoclasts, leukopenia, elevated serum levels of danger-associated molecular patterns (DAMPs; e.g. cell-free DNA and ATP) and cytokines (e.g. IL-1β and IL-18). Accordingly, doxorubicin activated the absent in melanoma (AIM2) and NLR family pyrin domain containing 3 (NLRP3) inflammasomes in macrophages and neutrophils, causing inflammatory cell death pyroptosis and NETosis, which correlated with its leukopenic effects. Moreover, the effects of this chemotherapeutic agent on cytokine secretion, cell demise, and bone loss were attenuated to various extent in conditions of AIM2 and/or NLRP3 insufficiency. Thus, we found that inflammasomes are key players in bone loss caused by doxorubicin, a finding that may inspire the development of a tailored adjuvant therapy that preserves the quality of this tissue in patients treated with this class of drugs.
Collapse
Affiliation(s)
- Chun Wang
- Division of Bone and Mineral Diseases, Washington University School of MedicineSt. LouisUnited States
| | - Khushpreet Kaur
- Division of Bone and Mineral Diseases, Washington University School of MedicineSt. LouisUnited States
| | - Canxin Xu
- Aclaris Therapeutics, IncSt. LouisUnited States
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of MedicineSt. LouisUnited States
- Shriners Hospitals for ChildrenSt. LouisUnited States
| | - Gabriel Mbalaviele
- Division of Bone and Mineral Diseases, Washington University School of MedicineSt. LouisUnited States
| |
Collapse
|
23
|
Lavelle EC, McEntee CP. Vaccine adjuvants: Tailoring innate recognition to send the right message. Immunity 2024; 57:772-789. [PMID: 38599170 DOI: 10.1016/j.immuni.2024.03.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
Adjuvants play pivotal roles in vaccine development, enhancing immunization efficacy through prolonged retention and sustained release of antigen, lymph node targeting, and regulation of dendritic cell activation. Adjuvant-induced activation of innate immunity is achieved via diverse mechanisms: for example, adjuvants can serve as direct ligands for pathogen recognition receptors or as inducers of cell stress and death, leading to the release of immunostimulatory-damage-associated molecular patterns. Adjuvant systems increasingly stimulate multiple innate pathways to induce greater potency. Increased understanding of the principles dictating adjuvant-induced innate immunity will subsequently lead to programming specific types of adaptive immune responses. This tailored optimization is fundamental to next-generation vaccines capable of inducing robust and sustained adaptive immune memory across different cohorts.
Collapse
Affiliation(s)
- Ed C Lavelle
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Craig P McEntee
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
24
|
Dvorkin S, Cambier S, Volkman HE, Stetson DB. New frontiers in the cGAS-STING intracellular DNA-sensing pathway. Immunity 2024; 57:718-730. [PMID: 38599167 PMCID: PMC11013568 DOI: 10.1016/j.immuni.2024.02.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 04/12/2024]
Abstract
The cGAS-STING intracellular DNA-sensing pathway has emerged as a key element of innate antiviral immunity and a promising therapeutic target. The existence of an innate immune sensor that can be activated by any double-stranded DNA (dsDNA) of any origin raises fundamental questions about how cGAS is regulated and how it responds to "foreign" DNA while maintaining tolerance to ubiquitous self-DNA. In this review, we summarize recent evidence implicating important roles for cGAS in the detection of foreign and self-DNA. We describe two recent and surprising insights into cGAS-STING biology: that cGAS is tightly tethered to the nucleosome and that the cGAMP product of cGAS is an immunotransmitter acting at a distance to control innate immunity. We consider how these advances influence our understanding of the emerging roles of cGAS in the DNA damage response (DDR), senescence, aging, and cancer biology. Finally, we describe emerging approaches to harness cGAS-STING biology for therapeutic benefit.
Collapse
Affiliation(s)
- Steve Dvorkin
- Departments of Immunology and Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Stephanie Cambier
- Departments of Immunology and Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Hannah E Volkman
- Departments of Immunology and Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Daniel B Stetson
- Departments of Immunology and Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
25
|
Ma M, Jiang W, Zhou R. DAMPs and DAMP-sensing receptors in inflammation and diseases. Immunity 2024; 57:752-771. [PMID: 38599169 DOI: 10.1016/j.immuni.2024.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/17/2024] [Accepted: 03/01/2024] [Indexed: 04/12/2024]
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous danger molecules produced in cellular damage or stress, and they can activate the innate immune system. DAMPs contain multiple types of molecules, including nucleic acids, proteins, ions, glycans, and metabolites. Although these endogenous molecules do not trigger immune response under steady-state condition, they may undergo changes in distribution, physical or chemical property, or concentration upon cellular damage or stress, and then they become DAMPs that can be sensed by innate immune receptors to induce inflammatory response. Thus, DAMPs play an important role in inflammation and inflammatory diseases. In this review, we summarize the conversion of homeostatic molecules into DAMPs; the diverse nature and classification, cellular origin, and sensing of DAMPs; and their role in inflammation and related diseases. Furthermore, we discuss the clinical strategies to treat DAMP-associated diseases via targeting DAMP-sensing receptors.
Collapse
Affiliation(s)
- Ming Ma
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Wei Jiang
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Rongbin Zhou
- Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China; Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
26
|
Vandepas LE, Stefani C, Domeier PP, Traylor-Knowles N, Goetz FW, Browne WE, Lacy-Hulbert A. Extracellular DNA traps in a ctenophore demonstrate immune cell behaviors in a non-bilaterian. Nat Commun 2024; 15:2990. [PMID: 38582801 PMCID: PMC10998917 DOI: 10.1038/s41467-024-46807-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 03/08/2024] [Indexed: 04/08/2024] Open
Abstract
The formation of extracellular DNA traps (ETosis) is a first response mechanism by specific immune cells following exposure to microbes. Initially characterized in vertebrate neutrophils, cells capable of ETosis have been discovered recently in diverse non-vertebrate taxa. To assess the conservation of ETosis between evolutionarily distant non-vertebrate phyla, we observed and quantified ETosis using the model ctenophore Mnemiopsis leidyi and the oyster Crassostrea gigas. Here we report that ctenophores - thought to have diverged very early from the metazoan stem lineage - possess immune-like cells capable of phagocytosis and ETosis. We demonstrate that both Mnemiopsis and Crassostrea immune cells undergo ETosis after exposure to diverse microbes and chemical agents that stimulate ion flux. We thus propose that ETosis is an evolutionarily conserved metazoan defense against pathogens.
Collapse
Affiliation(s)
- Lauren E Vandepas
- NRC Research Associateship Program, Seattle, WA, USA.
- Northwest Fisheries Science Center, National Oceanographic and Atmospheric Administration, Seattle, WA, 98112, USA.
- Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA.
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA.
| | - Caroline Stefani
- Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
| | - Phillip P Domeier
- Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
| | - Nikki Traylor-Knowles
- Rosenstiel School of Marine and Atmospheric Sciences, University of Miami, Miami, FL, 33149, USA
| | - Frederick W Goetz
- Northwest Fisheries Science Center, National Oceanographic and Atmospheric Administration, Seattle, WA, 98112, USA
| | - William E Browne
- Department of Biology, University of Miami, Coral Gables, FL, 33146, USA
| | - Adam Lacy-Hulbert
- Benaroya Research Institute at Virginia Mason, Seattle, WA, 98101, USA
| |
Collapse
|
27
|
Gewaid H, Bowie AG. Regulation of type I and type III interferon induction in response to pathogen sensing. Curr Opin Immunol 2024; 87:102424. [PMID: 38761566 DOI: 10.1016/j.coi.2024.102424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 02/19/2024] [Accepted: 05/06/2024] [Indexed: 05/20/2024]
Abstract
Type I and III interferons (IFN-I and IFN-III) have a central role in the early antimicrobial response against invading pathogens. Induction of IFN-Is and IFN-IIIs arises due to the sensing by pattern recognition receptors of pathogen-associated molecular patterns (from micro-organisms) or of damage-associated molecular patterns (DAMPs; produced by host cells). Here, we review recent developments on how IFN-I and IFN-III expression is stimulated by different pathogens and how the signalling pathways leading to IFN induction are tightly regulated. We also summarise the growing knowledge of the sensing pathways that lead to IFN-I and IFN-III induction in response to severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Hossam Gewaid
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew G Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
28
|
Oh T, Kang GS, Jo HJ, Park HJ, Lee YR, Ahn GO. DNA-dependent protein kinase regulates cytosolic double-stranded DNA secretion from irradiated macrophages to increase radiosensitivity of tumors. Radiother Oncol 2024; 193:110111. [PMID: 38286241 DOI: 10.1016/j.radonc.2024.110111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND AND PURPOSE To investigate the molecular mechanism by which irradiated macrophages secrete cytosolic double-stranded DNA (c-dsDNA) to increase radiosensitivity of tumors. MATERIALS AND METHODS Irradiated bone marrow-derived macrophages (BMDM) were co-incubated with irradiated EO771 or MC38 cancer cells to determine clonogenic survival. c-dsDNA were measured by agarose gel or enzyme-linked immunosorbent assay. BMDM or cancer cells were analyzed with immunostaining or western blot. Subcutaneously implanted MC38 cells in myeloid-specific Prkdc knockout (KO) mice or littermate control mice were irradiated with 8 Gy to determine radiosensitivity of tumors. RESULTS We observed that irradiated BMDM significantly increased radiosensitivity of cancer cells. By performing immunostaining, we found that there was a dose-dependent increase in the formation of c-dsDNA and phosphorylation in DNA-dependent protein kinase (DNA-PK) in irradiated BMDM. Importantly, c-dsDNA in irradiated BMDM could be secreted to the extracellular milieu and this process required DNA-PK, which phosphorylated myosin light chain to regulate the secretion. The secreted c-dsDNA from irradiated BMDM then activated toll-like receptor-9 and subsequent nuclear factor kappa-light-chain-enhancer of activated B cells signaling in the adjacent cancer cells inhibiting radiation-induced DNA double strand break repair. Lastly, we observed that irradiated tumors in vivo had a significantly increased number of tumor-associated macrophages (TAM) with phosphorylated DNA-PK expression in the cytosol. Furthermore, tumors grown in myeloid-specific Prkdc KO mice, in which TAM lacked phosphorylated DNA-PK expression were significantly more radioresistant than those of the wild-type control mice. CONCLUSIONS Irradiated macrophages can increase antitumor efficacy of radiotherapy through secretion of c-dsDNA under the regulation of DNA-PK.
Collapse
Affiliation(s)
- Taerim Oh
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Gi-Sue Kang
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Hye-Ju Jo
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Hye-Joon Park
- College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Ye-Rim Lee
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - G-One Ahn
- College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; College of Medicine, Seoul National University, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea.
| |
Collapse
|
29
|
Pan T, Lee JW. A crucial role of neutrophil extracellular traps in pulmonary infectious diseases. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2024; 2:34-41. [PMID: 39170960 PMCID: PMC11332830 DOI: 10.1016/j.pccm.2023.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Indexed: 08/23/2024]
Abstract
Neutrophil extracellular traps (NETs), extrusions of intracellular DNA with attached granular material that exert an antibacterial effect through entangling, isolating, and immobilizing microorganisms, have been extensively studied in recent decades. The primary role of NETs is to entrap and facilitate the killing of bacteria, fungi, viruses, and parasites, preventing bacterial and fungal dissemination. NET formation has been described in many pulmonary diseases, including both infectious and non-infectious. NETs are considered a double-edged sword. As innate immune cells, neutrophils release NETs to kill pathogens and remove cellular debris. However, the deleterious effects of excessive NET release in lung disease are particularly important because NETs and by-products of NETosis can directly induce epithelial and endothelial cell death while simultaneously inducing inflammatory cytokine secretion and immune-mediated thrombosis. Thus, NET formation must be tightly regulated to preserve the anti-microbial capability of NETs while minimizing damage to the host. In this review, we summarized the recent updates on the mechanism of NETs formation and pathophysiology associated with excessive NETs, aiming to provide insights for research and treatment of pulmonary infectious diseases.
Collapse
Affiliation(s)
- Ting Pan
- Shanghai Key Laboratory of Lung Inflammation and Injury, Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jae Woo Lee
- Department of Anesthesiology, University of California Los Angeles, Los Angeles, CA 90230, USA
| |
Collapse
|
30
|
Mousset A, Albrengues J. NETs unleashed: neutrophil extracellular traps boost chemotherapy against colorectal cancer. J Clin Invest 2024; 134:e178344. [PMID: 38426501 PMCID: PMC10904039 DOI: 10.1172/jci178344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024] Open
Abstract
Chemotherapy, which primarily acts on cancer cells, can influence the tumor microenvironment and the recruitment and behavior of stromal cells. In this issue of the JCI, Li et al. explored the potent anticancer effect of the combination of a glutaminase inhibitor (CB-839) and 5-FU against PIK3CA-mutant colorectal cancer tumors. This chemotherapy treatment strongly induced the recruitment of neutrophils that formed neutrophil extracellular traps in cancer, which actively killed cancer cells by inducing apoptosis. This study substantially advances our understanding of the multifaceted role of neutrophils and NETs in the outcome of anticancer treatment.
Collapse
|
31
|
Jhelum H, Čerina D, Harbort CJ, Lindner A, Hanitsch LG, Leistner R, Schröder JT, von Bernuth H, Stegemann MS, Schürmann M, Zychlinsky A, Krüger R, Marsman G. Panton-Valentine leukocidin-induced neutrophil extracellular traps lack antimicrobial activity and are readily induced in patients with recurrent PVL + -Staphylococcus aureus infections. J Leukoc Biol 2024; 115:222-234. [PMID: 37943843 DOI: 10.1093/jleuko/qiad137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 11/12/2023] Open
Abstract
Staphylococcus aureus strains that produce the toxin Panton-Valentine leukocidin (PVL-SA) frequently cause recurrent skin and soft tissue infections. PVL binds to and kills human neutrophils, resulting in the formation of neutrophil extracellular traps (NETs), but the pathomechanism has not been extensively studied. Furthermore, it is unclear why some individuals colonized with PVL-SA experience recurring infections whereas others are asymptomatic. We thus aimed to (1) investigate how PVL exerts its pathogenicity on neutrophils and (2) identify factors that could help to explain the predisposition of patients with recurring infections. We provide genetic and pharmacological evidence that PVL-induced NET formation is independent of NADPH oxidase and reactive oxygen species production. Moreover, through NET proteome analysis we identified that the protein content of PVL-induced NETs is different from NETs induced by mitogen or the microbial toxin nigericin. The abundance of the proteins cathelicidin (CAMP), elastase (NE), and proteinase 3 (PRTN3) was lower on PVL-induced NETs, and as such they were unable to kill S. aureus. Furthermore, we found that neutrophils from affected patients express higher levels of CD45, one of the PVL receptors, and are more susceptible to be killed at a low PVL concentration than control neutrophils. Neutrophils from patients that experience recurring PVL-positive infections may thus be more sensitive to PVL-induced NET formation, which might impair their ability to combat the infection.
Collapse
Affiliation(s)
- Hina Jhelum
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117, Berlin, Germany
| | - Dora Čerina
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117, Berlin, Germany
| | - C J Harbort
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117, Berlin, Germany
| | - Andreas Lindner
- Institute of Tropical Medicine and International Health, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Leif Gunnar Hanitsch
- Department of Medical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Rasmus Leistner
- Institute of Hygiene and Environmental Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Jennyver-Tabea Schröder
- Department of Pediatric Surgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Horst von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Department of Immunology, Labor Berlin GmbH, Sylter Straße 2, 13353, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Miriam Songa Stegemann
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Mariana Schürmann
- Department of Infectious Diseases and Respiratory Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Arturo Zychlinsky
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117, Berlin, Germany
| | - Renate Krüger
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117, Berlin, Germany
| | - Gerben Marsman
- Department of Cellular Microbiology, Max Planck Institute for Infection Biology, Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
32
|
Willemsen JF, Wenskus J, Lenz M, Rhode H, Trochimiuk M, Appl B, Pagarol-Raluy L, Börnigen D, Bang C, Reinshagen K, Herrmann M, Elrod J, Boettcher M. DNases improve effectiveness of antibiotic treatment in murine polymicrobial sepsis. Front Immunol 2024; 14:1254838. [PMID: 38259485 PMCID: PMC10801052 DOI: 10.3389/fimmu.2023.1254838] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 12/07/2023] [Indexed: 01/24/2024] Open
Abstract
Introduction Neutrophil extracellular traps (NETs) have various beneficial and detrimental effects in the body. It has been reported that some bacteria may evade the immune system when entangled in NETs. Thus, the aim of the current study was to evaluate the effects of a combined DNase and antibiotic therapy in a murine model of abdominal sepsis. Methods C57BL/6 mice underwent a cecum-ligation-and-puncture procedure. We used wild-type and knockout mice with the same genetic background (PAD4-KO and DNase1-KO). Mice were treated with (I) antibiotics (Metronidazol/Cefuroxime), (II) DNAse1, or (III) with the combination of both; mock-treated mice served as controls. We employed a streak plate procedure and 16s-RNA analysis to evaluate bacterial translocation and quantified NETs formation by ELISA and immune fluorescence. Western blot and proteomics analysis were used to determine inflammation. Results A total of n=73 mice were used. Mice that were genetically unable to produce extended NETs or were treated with DNases displayed superior survival and bacterial clearance and reduced inflammation. DNase1 treatment significantly improved clearance of Gram-negative bacteria and survival rates. Importantly, the combination of DNase1 and antibiotics reduced tissue damage, neutrophil activation, and NETs formation in the affected intestinal tissue. Conclusion The combination of antibiotics with DNase1 ameliorates abdominal sepsis. Gram-negative bacteria are cleared better when NETs are cleaved by DNase1. Future studies on antibiotic therapy should be combined with anti-NETs therapies.
Collapse
Affiliation(s)
- Jan-Fritjof Willemsen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Wenskus
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Moritz Lenz
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Holger Rhode
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Madgalena Trochimiuk
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Birgit Appl
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laia Pagarol-Raluy
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniela Börnigen
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Konrad Reinshagen
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Herrmann
- Department of Pediatric Surgery, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
- Department of Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitäts-klinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich Alexander University Erlangen-Nuremberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Elrod
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Surgery, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Surgery, University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
33
|
Li Y, Wu Y, Huang J, Cao X, An Q, Peng Y, Zhao Y, Luo Y. A variety of death modes of neutrophils and their role in the etiology of autoimmune diseases. Immunol Rev 2024; 321:280-299. [PMID: 37850797 DOI: 10.1111/imr.13284] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Neutrophils are important in the context of innate immunity and actively contribute to the progression of diverse autoimmune disorders. Distinct death mechanisms of neutrophils may exhibit specific and pivotal roles in autoimmune diseases and disease pathogenesis through the orchestration of immune homeostasis, the facilitation of autoantibody production, the induction of tissue and organ damage, and the incitement of pathological alterations. In recent years, more studies have provided in-depth examination of various neutrophil death modes, revealing nuances that challenge conventional understanding and underscoring their potential clinical utility in diagnosis and treatment. This review explores the multifaceted processes and characteristics of neutrophil death, with a focus on tailored investigations within various autoimmune diseases. It also highlights the potential interplay between neutrophil death and the landscape of autoimmune disorders. The review encapsulates the pertinent pathways implicated in various neutrophil death mechanisms across diverse autoimmune diseases while also charts possible avenues for future research.
Collapse
Affiliation(s)
- Yanhong Li
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yinlan Wu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingang Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xue Cao
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Qiyuan An
- School of Inspection and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yun Peng
- Department of Rheumatology and Clinical Immunology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Yi Zhao
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yubin Luo
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
34
|
Shafqat A, Khan JA, Alkachem AY, Sabur H, Alkattan K, Yaqinuddin A, Sing GK. How Neutrophils Shape the Immune Response: Reassessing Their Multifaceted Role in Health and Disease. Int J Mol Sci 2023; 24:17583. [PMID: 38139412 PMCID: PMC10744338 DOI: 10.3390/ijms242417583] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Neutrophils are the most abundant of the circulating immune cells and are the first to be recruited to sites of inflammation. Neutrophils are a heterogeneous group of immune cells from which are derived extracellular traps (NETs), reactive oxygen species, cytokines, chemokines, immunomodulatory factors, and alarmins that regulate the recruitment and phenotypes of neutrophils, macrophages, dendritic cells, T cells, and B cells. In addition, cytokine-stimulated neutrophils can express class II major histocompatibility complex and the internal machinery necessary for successful antigen presentation to memory CD4+ T cells. This may be relevant in the context of vaccine memory. Neutrophils thus emerge as orchestrators of immune responses that play a key role in determining the outcome of infections, vaccine efficacy, and chronic diseases like autoimmunity and cancer. This review aims to provide a synthesis of current evidence as regards the role of these functions of neutrophils in homeostasis and disease.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia (K.A.); (A.Y.); (G.K.S.)
| | | | | | | | | | | | | |
Collapse
|
35
|
Zhou Z, Ou-yang C, Chen Q, Ren Z, Guo X, Lei M, Liu C, Yang X. Trafficking and effect of released DNA on cGAS-STING signaling pathway and cardiovascular disease. Front Immunol 2023; 14:1287130. [PMID: 38152400 PMCID: PMC10751357 DOI: 10.3389/fimmu.2023.1287130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/01/2023] [Indexed: 12/29/2023] Open
Abstract
Evidence from clinical research and animal studies indicates that inflammation is an important factor in the occurrence and development of cardiovascular disease (CVD). Emerging evidence shows that nucleic acids serve as crucial pathogen-associated molecular patterns (PAMPs) or non-infectious damage-associated molecular patterns (DAMPs), are released and then recognized by pattern recognition receptors (PRRs), which activates immunological signaling pathways for host defense. Mechanistically, the released nucleic acids activate cyclic GMP-AMP synthase (cGAS) and its downstream receptor stimulator of interferon genes (STING) to promote type I interferons (IFNs) production, which play an important regulatory function during the initiation of an innate immune response to various diseases, including CVD. This pathway represents an essential defense regulatory mechanism in an organism's innate immune system. In this review, we outline the overall profile of cGAS-STING signaling, summarize the latest findings on nucleic acid release and trafficking, and discuss their potential role in CVD. This review also sheds light on potential directions for future investigations on CVD.
Collapse
Affiliation(s)
- Zimo Zhou
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
- State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Changhan Ou-yang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qingjie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhanhong Ren
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Xiying Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Min Lei
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Xiaosong Yang
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, China
- Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
36
|
Liu Y, Wang R, Song C, Ding S, Zuo Y, Yi K, Li N, Wang B, Geng Q. Crosstalk between neutrophil extracellular traps and immune regulation: insights into pathobiology and therapeutic implications of transfusion-related acute lung injury. Front Immunol 2023; 14:1324021. [PMID: 38162674 PMCID: PMC10755469 DOI: 10.3389/fimmu.2023.1324021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Transfusion-related acute lung injury (TRALI) is the leading cause of transfusion-associated death, occurring during or within 6 hours after transfusion. Reports indicate that TRALI can be categorized as having or lacking acute respiratory distress syndrome (ARDS) risk factors. There are two types of TRALI in terms of its pathogenesis: antibody-mediated and non-antibody-mediated. The key initiation steps involve the priming and activation of neutrophils, with neutrophil extracellular traps (NETs) being established as effector molecules formed by activated neutrophils in response to various stimuli. These NETs contribute to the production and release of reactive oxygen species (ROS) and participate in the destruction of pulmonary vascular endothelial cells. The significant role of NETs in TRALI is well recognized, offering a potential pathway for TRALI treatment. Moreover, platelets, macrophages, endothelial cells, and complements have been identified as promoters of NET formation. Concurrently, studies have demonstrated that the storage of platelets and concentrated red blood cells (RBC) can induce TRALI through bioactive lipids. In this article, recent clinical and pre-clinical studies on the pathophysiology and pathogenesis of TRALI are reviewed to further illuminate the mechanism through which NETs induce TRALI. This review aims to propose new therapeutic strategies for TRALI, with the hope of effectively improving its poor prognosis.
Collapse
Affiliation(s)
- Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Rong Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Congkuan Song
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yifan Zuo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Yi
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bo Wang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
37
|
Kubota T. An Emerging Role for Anti-DNA Antibodies in Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:16499. [PMID: 38003689 PMCID: PMC10671047 DOI: 10.3390/ijms242216499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Anti-DNA antibodies are hallmark autoantibodies produced in systemic lupus erythematosus (SLE), but their pathogenetic role is not fully understood. Accumulating evidence suggests that some anti-DNA antibodies enter different types of live cells and affect the pathophysiology of SLE by stimulating or impairing these cells. Circulating neutrophils in SLE are activated by a type I interferon or other stimuli and are primed to release neutrophil extracellular traps (NETs) on additional stimulation. Anti-DNA antibodies are also involved in this process and may induce NET release. Thereafter, they bind and protect extracellular DNA in the NETs from digestion by nucleases, resulting in increased NET immunogenicity. This review discusses the pathogenetic role of anti-DNA antibodies in SLE, mainly focusing on recent progress in the two research fields concerning antibody penetration into live cells and NETosis.
Collapse
Affiliation(s)
- Tetsuo Kubota
- Department of Medical Technology, Tsukuba International University, Tsuchiura 300-0051, Ibaraki, Japan
| |
Collapse
|
38
|
Chang PW, Wang JY, Wang WP, Huang WC, Wu MH, Song JS, Chen LY, Tung CW, Chi YH, Ueng SH. Analysis of structure-activity relationship of indol-3-yl-N-phenylcarbamic amides as potent STING inhibitors. Bioorg Med Chem 2023; 95:117502. [PMID: 37866089 DOI: 10.1016/j.bmc.2023.117502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/05/2023] [Accepted: 10/13/2023] [Indexed: 10/24/2023]
Abstract
A structure-activity relationship (SAR) study of stimulator of interferon gene (STING) inhibition was performed using a series of indol-3-yl-N-phenylcarbamic amides and indol-2-yl-N-phenylcarbamic amides. Among these analogs, compounds 10, 13, 15, 19, and 21 inhibited the phosphorylation of STING and interferon regulatory factor 3 (IRF3) to a greater extent than the reference compound, H-151. All five analogs showed stronger STING inhibition than H-151 on the 2',3'-cyclic GMP-AMP-induced expression of interferon regulatory factors (IRFs) in a STINGR232 knock-in THP-1 reporter cell line. The half-maximal inhibitory concentration of the most potent compound, 21, was 11.5 nM. The molecular docking analysis of compound 21 and STING combined with the SAR study suggested that the meta- and para-positions of the benzene ring of the phenylcarbamic amide moiety could be structurally modified by introducing halides or alkyl substituents.
Collapse
Affiliation(s)
- Po-Wei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC
| | - Jing-Ya Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC
| | - Wan-Ping Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC
| | - Wei-Cheng Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC
| | - Mine-Hsine Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC
| | - Liuh-Yow Chen
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan, ROC
| | - Chun-Wei Tung
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC
| | - Ya-Hui Chi
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC.
| | - Shau-Hua Ueng
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan, ROC; School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan, ROC.
| |
Collapse
|
39
|
García-Giménez J, Córdoba-David G, Rayego-Mateos S, Cannata-Ortiz P, Carrasco S, Ruiz-Ortega M, Fernandez-Fernandez B, Ortiz A, Ramos AM. STING1 deficiency ameliorates immune-mediated crescentic glomerulonephritis in mice. J Pathol 2023; 261:309-322. [PMID: 37650295 DOI: 10.1002/path.6177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/10/2023] [Accepted: 07/20/2023] [Indexed: 09/01/2023]
Abstract
Rapidly progressive/crescentic glomerulonephritis (RPGN/CGN) involves the formation of glomerular crescents by maladaptive differentiation of parietal epithelial cells that leads to rapid loss of renal function. The molecular mechanisms of crescent formation are poorly understood. Therefore, new insights into molecular mechanisms could identify alternative therapeutic targets for RPGN/CGN. Analysis of kidney biopsies from patients with RPGN revealed increased interstitial, glomerular, and tubular expression of STING1, an accessory protein of the c-GAS-dependent DNA-sensing pathway, which was also observed in murine nephrotoxic nephritis induced by an anti-GBM antibody. STING1 was expressed by key cell types involved in RPGN and crescent formation such as glomerular parietal epithelial cells, and tubular cells as well as by inflammation accessory cells. In functional in vivo studies, Sting1-/- mice with nephrotoxic nephritis had lower kidney cytokine expression, milder kidney infiltration by innate and adaptive immune cells, and decreased disease severity. Pharmacological STING1 inhibition mirrored these findings. Direct STING1 agonism in parietal and tubular cells activated the NF-κB-dependent cytokine response and the interferon-induced genes (ISGs) program. These responses were also triggered in a STING1-dependent manner by the pro-inflammatory cytokine TWEAK. These results identify STING1 activation as a pathological mechanism in RPGN/CGN and TWEAK as an activator of STING1. Pharmacological strategies targeting STING1, or upstream regulators may therefore be potential alternatives to treat RPGN. © 2023 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jorge García-Giménez
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Gina Córdoba-David
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Pablo Cannata-Ortiz
- Department of Pathology, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Susana Carrasco
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Beatriz Fernandez-Fernandez
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medicine, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Ortiz
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Pharmacology, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
| | - Adrián M Ramos
- Department of Nephrology and Hypertension, IIS-Fundación Jiménez Díaz-Universidad Autónoma de Madrid, Madrid, Spain
- RICORS2040, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW Platelet factor 4 (PF4, CXCL4), the most abundant α-granule platelet-specific chemokine, forms tetramers with an equatorial ring of high positive charge that bind to a wide range of polyanions, after which it changes conformation to expose antigenic epitopes. Antibodies directed against PF4 not only help to clear infection but can also lead to the development of thrombotic disorders such as heparin-induced thrombocytopenia (HIT) and vaccine-induced thrombocytopenia and thrombosis (VITT). This review will outline the different mechanisms through which PF4 engagement with polyanions combats infection but also contributes to the pathogenesis of inflammatory and thrombotic disease states. RECENT FINDINGS Recent work has shown that PF4 binding to microbial polyanions may improve outcomes in infection by enhancing leukocyte-bacterial binding, tethering pathogens to neutrophil extracellular traps (NETs), decreasing the thrombotic potential of NET DNA, and modulating viral infectivity. However, PF4 binding to nucleic acids may enhance their recognition by innate immune receptors, leading to autoinflammation. Lastly, while HIT is induced by platelet activating antibodies that bind to PF4/polyanion complexes, VITT, which occurs in a small subset of patients treated with COVID-19 adenovirus vector vaccines, is characterized by prothrombotic antibodies that bind to PF4 alone. SUMMARY Investigating the complex interplay of PF4 and polyanions may provide insights relevant to the treatment of infectious disease while also improving our understanding of the pathogenesis of thrombotic disorders driven by anti-PF4/polyanion and anti-PF4 antibodies.
Collapse
Affiliation(s)
- Anh T P Ngo
- Division of Hematology, Children's Hospital of Philadelphia
| | | | - Kandace Gollomp
- Division of Hematology, Children's Hospital of Philadelphia
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
41
|
Sun C, Shi H, Zhao X, Chang YL, Wang X, Zhu S, Sun S. The Activation of cGAS-STING in Acute Kidney Injury. J Inflamm Res 2023; 16:4461-4470. [PMID: 37842189 PMCID: PMC10576462 DOI: 10.2147/jir.s423232] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/01/2023] [Indexed: 10/17/2023] Open
Abstract
The activation of the cGAS-STING pathway is associated with many sterile inflammatory and inflammatory conditions, including acute kidney injury. As a cytoplasmic DNA sensor, sensitization of the cGAS-STING pathway can ignite the innate immune response in vivo and trigger a series of biological effects. In recent years, there is increasing evidence showing that the cGAS-STING pathway plays a vital role in acute kidney injury, a non-inflammatory disease induced by activation of innate immune cells, and closely related to intracellular reactive oxygen species, mitochondrial DNA, and the cGAS-STING pathway. This review provides a prospect of the cGAS-STING pathway and its relationship to acute kidney injury.
Collapse
Affiliation(s)
- Chuanchuan Sun
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Heng Shi
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Xinhai Zhao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Yu-Ling Chang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Xianghong Wang
- Department of Endocrinology and Metabolism, Zhuhai Hospital Affiliated with Jinan University (Zhuhai People’s Hospital), Zhuhai, People’s Republic of China
| | - Shiping Zhu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| | - Shengyun Sun
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, People’s Republic of China
| |
Collapse
|
42
|
Shrestha S, Hong CW. Extracellular Mechanisms of Neutrophils in Immune Cell Crosstalk. Immune Netw 2023; 23:e38. [PMID: 37970234 PMCID: PMC10643328 DOI: 10.4110/in.2023.23.e38] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/24/2023] [Accepted: 09/05/2023] [Indexed: 11/17/2023] Open
Abstract
Neutrophils are professional phagocytes that provide defense against invading pathogens through phagocytosis, degranulation, generation of ROS, and the formation of neutrophil extracellular traps (NETs). Although long been considered as short-lived effector cells with limited biosynthetic activity, recent studies have revealed that neutrophils actively communicate with other immune cells. Neutrophils employ various types of soluble mediators, including granules, cytokines, and chemokines, for crosstalk with immune cells. Additionally, ROS and NETs, major arsenals of neutrophils, are utilized for intercellular communication. Furthermore, extracellular vesicles play a crucial role as mediators of neutrophil crosstalk. In this review, we highlight the extracellular mechanisms of neutrophils and their roles in crosstalk with other cells.
Collapse
Affiliation(s)
- Sanjeeb Shrestha
- Department of Physiology, CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Chang-Won Hong
- Department of Physiology, CMRI, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
43
|
Mayer-Barber KD. Granulocytes subsets and their divergent functions in host resistance to Mycobacterium tuberculosis - a 'tipping-point' model of disease exacerbation. Curr Opin Immunol 2023; 84:102365. [PMID: 37437471 PMCID: PMC10543468 DOI: 10.1016/j.coi.2023.102365] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/29/2023] [Accepted: 06/14/2023] [Indexed: 07/14/2023]
Abstract
Granulocytes are innate immune effector cells with essential functions in host resistance to bacterial infections. I will discuss emerging evidence that during Mycobacterium tuberculosis infection, counter-intuitively, eosinophils are host-protective while neutrophils are host detrimental. Additionally, I will propose a 'tipping-point' model in which neutrophils are an integral part of a feedforward loop driving tuberculosis disease exacerbation.
Collapse
Affiliation(s)
- Katrin D Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, 20892, USA.
| |
Collapse
|
44
|
Shi G, Liu L, Cao Y, Ma G, Zhu Y, Xu J, Zhang X, Li T, Mi L, Jia H, Zhang Y, Liu X, Zhou Y, Li S, Yang G, Liu X, Chen F, Wang B, Deng Q, Zhang S, Zhang J. Inhibition of neutrophil extracellular trap formation ameliorates neuroinflammation and neuronal apoptosis via STING-dependent IRE1α/ASK1/JNK signaling pathway in mice with traumatic brain injury. J Neuroinflammation 2023; 20:222. [PMID: 37777772 PMCID: PMC10543875 DOI: 10.1186/s12974-023-02903-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/22/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Neuroinflammation is one of the most important pathogeneses in secondary brain injury after traumatic brain injury (TBI). Neutrophil extracellular traps (NETs) forming neutrophils were found throughout the brain tissue of TBI patients and elevated plasma NET biomarkers correlated with worse outcomes. However, the biological function and underlying mechanisms of NETs in TBI-induced neural damage are not yet fully understood. Here, we used Cl-amidine, a selective inhibitor of NETs to investigate the role of NETs in neural damage after TBI. METHODS Controlled cortical impact model was performed to establish TBI. Cl-amidine, 2'3'-cGAMP (an activator of stimulating Interferon genes (STING)), C-176 (a selective STING inhibitor), and Kira6 [a selectively phosphorylated inositol-requiring enzyme-1 alpha [IRE1α] inhibitor] were administrated to explore the mechanism by which NETs promote neuroinflammation and neuronal apoptosis after TBI. Peptidyl arginine deiminase 4 (PAD4), an essential enzyme for neutrophil extracellular trap formation, is overexpressed with adenoviruses in the cortex of mice 1 day before TBI. The short-term neurobehavior tests, magnetic resonance imaging (MRI), laser speckle contrast imaging (LSCI), Evans blue extravasation assay, Fluoro-Jade C (FJC), TUNEL, immunofluorescence, enzyme-linked immunosorbent assay (ELISA), western blotting, and quantitative-PCR were performed in this study. RESULTS Neutrophils form NETs presenting in the circulation and brain at 3 days after TBI. NETs inhibitor Cl-amidine treatment improved short-term neurological functions, reduced cerebral lesion volume, reduced brain edema, and restored cerebral blood flow (CBF) after TBI. In addition, Cl-amidine exerted neuroprotective effects by attenuating BBB disruption, inhibiting immune cell infiltration, and alleviating neuronal death after TBI. Moreover, Cl-amidine treatment inhibited microglia/macrophage pro-inflammatory polarization and promoted anti-inflammatory polarization at 3 days after TBI. Mechanistically, STING ligand 2'3'-cGAMP abolished the neuroprotection of Cl-amidine via IRE1α/ASK1/JNK signaling pathway after TBI. Importantly, overexpression of PAD4 promotes neuroinflammation and neuronal death via the IRE1α/ASK1/JNK signaling pathway after TBI. However, STING inhibitor C-176 or IRE1α inhibitor Kira6 effectively abolished the neurodestructive effects of PAD4 overexpression after TBI. CONCLUSION Altogether, we are the first to demonstrate that NETs inhibition with Cl-amidine ameliorated neuroinflammation, neuronal apoptosis, and neurological deficits via STING-dependent IRE1α/ASK1/JNK signaling pathway after TBI. Thus, Cl-amidine treatment may provide a promising therapeutic approach for the early management of TBI.
Collapse
Affiliation(s)
- Guihong Shi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Liang Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Yiyao Cao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Guangshuo Ma
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
- Department of Neurosurgery, School of Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300192, China
| | - Yanlin Zhu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Jianye Xu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Xu Zhang
- School of Medicine, Nankai University, Tianjin, 300192, China
| | - Tuo Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Liang Mi
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Haoran Jia
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Yanfeng Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Xilei Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Yuan Zhou
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Shenghui Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Guili Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Xiao Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Fanglian Chen
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Baolong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Quanjun Deng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China
| | - Shu Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China.
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
- Key Laboratory of Post-Trauma Neuro-Repair and Regeneration in Central Nervous System, Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, 300052, People's Republic of China.
| |
Collapse
|
45
|
Sun S, Lv W, Li S, Zhang Q, He W, Min Z, Teng C, Chen Y, Liu L, Yin J, Zhu B, Xu M, Dai D, Xin H. Smart Liposomal Nanocarrier Enhanced the Treatment of Ischemic Stroke through Neutrophil Extracellular Traps and Cyclic Guanosine Monophosphate-Adenosine Monophosphate Synthase-Stimulator of Interferon Genes (cGAS-STING) Pathway Inhibition of Ischemic Penumbra. ACS NANO 2023; 17:17845-17857. [PMID: 37712845 DOI: 10.1021/acsnano.3c03390] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
Brain inflammation is regarded as one of the leading causes that aggravates secondary brain injury and hinders the prognosis of ischemic stroke. After ischemic stroke, high quantities of peripheral neutrophils are recruited to brain lesions and release neutrophil extracellular traps (NETs), leading to the aggravation of blood-brain barrier (BBB) damage, activation of microglia, and ultimate neuronal death. Herein, a smart multifunctional delivery system has been developed to regulate immune disorders in the ischemic brain. Briefly, Cl-amidine, an inhibitor of peptidylarginine deiminase 4 (PAD4), is encapsulated into self-assembled liposomal nanocarriers (C-Lipo/CA) that are modified by reactive oxygen species (ROS)-responsive polymers and fibrin-binding peptide to achieve targeting ischemic lesions and stimuli-responsive release of a drug. In the mouse model of cerebral artery occlusion/reperfusion (MCAO), C-Lipo/CA can suppress the NETs release process (NETosis) and further inhibit the cyclic guanosine monophosphate-adenosine monophosphate synthase-stimulator of interferon genes (cGAS-STING) pathway in an ischemic brain. In addition, MCAO mice treated with C-Lipo/CA significantly mitigated ischemic and reperfusion injury, with a reduction in the area of cerebral infarction to 12.1%, compared with the saline group of about 46.7%. These results demonstrated that C-Lipo/CA, which integrated microglia regulation, BBB protection, and neuron survival, exerts a potential therapy strategy to maximize ameliorating the mortality of ischemic stroke.
Collapse
Affiliation(s)
- Shanbo Sun
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Wei Lv
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214400, China
| | - Shengnan Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qi Zhang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Weichong He
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zhiyi Min
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chuanhui Teng
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuqin Chen
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Linfeng Liu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jiaqing Yin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Baoli Zhu
- Jiangsu Engineering Research Center of Health Emergency, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ming Xu
- Jiangsu Engineering Research Center of Health Emergency, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
- School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Dongwei Dai
- Department of Neurosurgery, The First Affiliated Hospital of Naval Medical University, Changhai Hospital of Shanghai, Shanghai 200433, China
| | - Hongliang Xin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
46
|
Manda-Handzlik A, Cieloch A, Kuźmicka W, Mroczek A, Stelmaszczyk-Emmel A, Demkow U, Wachowska M. Secretomes of M1 and M2 macrophages decrease the release of neutrophil extracellular traps. Sci Rep 2023; 13:15633. [PMID: 37730741 PMCID: PMC10511515 DOI: 10.1038/s41598-023-42167-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/06/2023] [Indexed: 09/22/2023] Open
Abstract
The release of neutrophil extracellular traps (NETs) can be either beneficial or detrimental for the host, thus it is necessary to maintain a balance between formation and clearance of NETs. Multiple physiological factors eliciting NET release have been identified, yet the studies on natural signals limiting NET formation have been scarce. Accordingly, our aim was to analyze whether cytokines or immune cells can inhibit NET formation. To that end, human granulocytes were incubated with interleukin (IL)-4, IL-10, transforming growth factor beta-2 or adenosine and then stimulated to release NETs. Additionally, neutrophils were cultured in the presence of natural killer (NK) cells, regulatory T cells (Tregs), pro-inflammatory or anti-inflammatory macrophages (M1 or M2 macrophages), or in the presence of NK/Tregs/M1 macrophages or M2 macrophages-conditioned medium and subsequently stimulated to release NETs. Our studies showed that secretome of M1 and M2 macrophages, but not of NK cells and Tregs, diminishes NET formation. Co-culture experiments did not reveal any effect of immune cells on NET release. No effect of cytokines or adenosine on NET release was found. This study highlights the importance of paracrine signaling at the site of infection and is the first to show that macrophage secretome can regulate NET formation.
Collapse
Affiliation(s)
- Aneta Manda-Handzlik
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland.
| | - Adrianna Cieloch
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091, Warsaw, Poland
| | - Weronika Kuźmicka
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| | - Agnieszka Mroczek
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
- Doctoral School, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091, Warsaw, Poland
| | - Anna Stelmaszczyk-Emmel
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| | - Urszula Demkow
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| | - Małgorzata Wachowska
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| |
Collapse
|
47
|
Zhu S, Yu Y, Qu M, Qiu Z, Zhang H, Miao C, Guo K. Neutrophil extracellular traps contribute to immunothrombosis formation via the STING pathway in sepsis-associated lung injury. Cell Death Discov 2023; 9:315. [PMID: 37626060 PMCID: PMC10457383 DOI: 10.1038/s41420-023-01614-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/04/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are involved in the activation and dysfunction of multiple overlapping and interacting pathways, including the immune response to injury, inflammation, and coagulation, which contribute to the pathogenesis of sepsis-induced acute lung injury (SI-ALI). However, how NETs mediate the relationship between inflammation and coagulation has not been fully clarified. Here, we found that NETs, through stimulator of interferon genes (STING) activation, induced endothelial cell damage with abundant production of tissue factor (TF), which magnified the dysregulation between inflammatory and coagulant responses and resulted in poor prognosis of SI-ALI model mice. Disruption of NETs and inhibition of STING improved the outcomes of septic mice and reduced the inflammatory response and coagulation. Furthermore, Toll-like receptor 2 (TLR2) on the surface of endothelial cells was involved in the interaction between NETs and the STING pathway. Collectively, these findings demonstrate that NETs activate the coagulant cascade in endothelial cells in a STING-dependent manner in the development of SI-ALI.
Collapse
Affiliation(s)
- Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhiyun Qiu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
48
|
Melbouci D, Haidar Ahmad A, Decker P. Neutrophil extracellular traps (NET): not only antimicrobial but also modulators of innate and adaptive immunities in inflammatory autoimmune diseases. RMD Open 2023; 9:e003104. [PMID: 37562857 PMCID: PMC10423839 DOI: 10.1136/rmdopen-2023-003104] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 05/14/2023] [Indexed: 08/12/2023] Open
Abstract
Polymorphonuclear neutrophils (PMN) represent one of the first lines of defence against invading pathogens and are the most abundant leucocytes in the circulation. Generally described as pro-inflammatory cells, recent data suggest that PMN also have immunomodulatory capacities. In response to certain stimuli, activated PMN expel neutrophil extracellular traps (NET), structures made of DNA and associated proteins. Although originally described as an innate immune mechanism fighting bacterial infection, NET formation (or probably rather an excess of NET together with impaired clearance of NET) may be deleterious. Indeed, NET have been implicated in the development of several inflammatory and autoimmune diseases as rheumatoid arthritis or systemic lupus erythematosus, as well as fibrosis or cancer. They have been suggested as a source of (neo)autoantigens or regulatory proteins like proteases or to act as a physical barrier. Different mechanisms of NET formation have been described, leading to PMN death or not, depending on the stimulus. Interestingly, NET may be both pro-inflammatory and anti-inflammatory and this probably partly depends on the mechanism, and thus the stimuli, triggering NET formation. Within this review, we will describe the pro-inflammatory and anti-inflammatory activities of NET and especially how NET may modulate immune responses.
Collapse
Affiliation(s)
- Dyhia Melbouci
- Inserm UMR 1125, Li2P, Université Sorbonne Paris Nord-Campus de Bobigny, Bobigny, Île-de-France, France
| | - Ahmad Haidar Ahmad
- Inserm UMR 1125, Li2P, Université Sorbonne Paris Nord-Campus de Bobigny, Bobigny, Île-de-France, France
| | - Patrice Decker
- Inserm UMR 1125, Li2P, Université Sorbonne Paris Nord-Campus de Bobigny, Bobigny, Île-de-France, France
| |
Collapse
|
49
|
Mankan AK, Czajka-Francuz P, Prendes M, Ramanan S, Koziej M, Vidal L, Saini KS. Intracellular DNA sensing by neutrophils and amplification of the innate immune response. Front Immunol 2023; 14:1208137. [PMID: 37483598 PMCID: PMC10361817 DOI: 10.3389/fimmu.2023.1208137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/13/2023] [Indexed: 07/25/2023] Open
Abstract
As the first responders, neutrophils lead the innate immune response to infectious pathogens and inflammation inducing agents. The well-established pathogen neutralizing strategies employed by neutrophils are phagocytosis, the action of microbicide granules, the production of ROS, and the secretion of neutrophil extracellular traps (NETs). Only recently, the ability of neutrophils to sense and respond to pathogen-associated molecular patterns is being appreciated. This review brings together the current information about the intracellular recognition of DNA by neutrophils and proposes models of signal amplification in immune response. Finally, the clinical relevance of DNA sensing by neutrophils in infectious and non-infectious diseases including malignancy are also discussed.
Collapse
Affiliation(s)
| | | | - Maria Prendes
- Labcorp Drug Development Inc., Princeton, NJ, United States
| | - Sriram Ramanan
- Labcorp Drug Development Inc., Princeton, NJ, United States
| | | | | | - Kamal S. Saini
- Fortrea, Inc., Durham, NC, United States
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
50
|
Fritsch LE, Kelly C, Pickrell AM. The role of STING signaling in central nervous system infection and neuroinflammatory disease. WIREs Mech Dis 2023; 15:e1597. [PMID: 36632700 PMCID: PMC10175194 DOI: 10.1002/wsbm.1597] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/27/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023]
Abstract
The cyclic guanosine monophosphate-adenosine monophosphate (GMP-AMP) synthase-Stimulator of Interferon Genes (cGAS-STING) pathway is a critical innate immune mechanism for detecting the presence of double-stranded DNA (dsDNA) and prompting a robust immune response. Canonical cGAS-STING activation occurs when cGAS, a predominantly cytosolic pattern recognition receptor, binds microbial DNA to promote STING activation. Upon STING activation, transcription factors enter the nucleus to cause the production of Type I interferons, inflammatory cytokines whose primary function is to prime the host for viral infection by producing a number of antiviral interferon-stimulated genes. While the pathway was originally described in viral infection, more recent studies have implicated cGAS-STING signaling in a number of different contexts, including autoimmune disease, cancer, injury, and neuroinflammatory disease. This review focuses on how our understanding of the cGAS-STING pathway has evolved over time with an emphasis on the role of STING-mediated neuroinflammation and infection in the nervous system. We discuss recent findings on how STING signaling contributes to the pathology of pain, traumatic brain injury, and stroke, as well as how mitochondrial DNA may promote STING activation in common neurodegenerative diseases. We conclude by commenting on the current knowledge gaps that should be filled before STING can be an effective therapeutic target in neuroinflammatory disease. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology Infectious Diseases > Molecular and Cellular Physiology Immune System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Lauren E. Fritsch
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Roanoke, Virginia, USA
| | - Colin Kelly
- Graduate Program in Translational Biology, Medicine, and Health, Virginia Polytechnic Institute and State University, Roanoke, Virginia, USA
| | - Alicia M. Pickrell
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| |
Collapse
|