351
|
Hounkpe BW, Chenou F, Domingos IDF, Cardoso EC, Costa Sobreira MJDV, Araujo AS, Lucena‐Araújo AR, da Silva Neto PV, Malheiro A, Fraiji NA, Costa FF, Bezerra MAC, Santos MNN, De Paula EV. Neutrophil extracellular trap regulators in sickle cell disease: Modulation of gene expression of PADI4, neutrophil elastase, and myeloperoxidase during vaso-occlusive crisis. Res Pract Thromb Haemost 2021; 5:204-210. [PMID: 33537545 PMCID: PMC7845058 DOI: 10.1002/rth2.12463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 10/28/2020] [Accepted: 10/31/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent evidence suggests that generation of neutrophil extracellular traps (NETosis), one of the components of immunothrombosis, is associated with the pathogenesis of both venous thromboembolism and sickle cell disease (SCD). NETosis is a complex process regulated by several proteins such as peptidyl arginine deaminase 4 (PADI4), neutrophil elastase (ELANE), and myeloperoxidase (MPO). Among these regulators, PADI4 is responsible of histone citrullination, an essential step for NETosis. Accordingly, its inhibition has been recently cited as a promising therapeutic strategy for diseases such as SCD. Although attractive, this strategy requires supportive evidence of its role in the pathogenesis of SCD. PATIENTS AND METHODS Patients from two independent cohorts were enrolled in this study. Samples were obtained at steady state (53 patients) or during acute episodes of vaso-occlusive crisis (VOC; 28 patients) in patients from cohort 1. mRNA was extracted from granulocytes to analyze PADI4, ELANE, and MPO expression by qPCR. Furthermore, plasma activity of PADI4 was assessed from an independent cohort in 15 patients, within 24 hours from admission for VOC. Race-matched healthy individuals from the same geographic regions were used as controls for each cohort. RESULTS AND CONCLUSIONS Higher levels of gene expression of PADI4 and ELANE were observed during VOC. Furthermore, plasma activity of PADI4 was higher in acute VOC when compared to healthy individuals. These results demonstrate that NETosis regulators are modulated during acute VOC, and pave the way for studies of PADI4 inhibition as a therapeutic strategy for acute VOC in SCD.
Collapse
Affiliation(s)
| | - Francine Chenou
- School of Medical SciencesUniversity of CampinasCampinasBrazil
| | | | | | | | - Aderson S. Araujo
- Hematology and Hemotherapy Foundation of Pernambuco ‐ HEMOPERecifeBrazil
| | | | | | - Adriana Malheiro
- Hematology and Hemotherapy Foundation from Amazonas State (HEMOAM)ManausBrazil
| | | | - Fernando Ferreira Costa
- School of Medical SciencesUniversity of CampinasCampinasBrazil
- Hematology and Hemotherapy CenterUniversity of CampinasCampinasBrazil
| | | | | | - Erich Vinicius De Paula
- School of Medical SciencesUniversity of CampinasCampinasBrazil
- Hematology and Hemotherapy CenterUniversity of CampinasCampinasBrazil
| |
Collapse
|
352
|
Kuhikar R, Khan N, Khare SP, Fulzele A, Melinkeri S, Kale V, Limaye L. Neutrophils generated in vitro from hematopoietic stem cells isolated from apheresis samples and umbilical cord blood form neutrophil extracellular traps. Stem Cell Res 2020; 50:102150. [PMID: 33450673 DOI: 10.1016/j.scr.2020.102150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 11/05/2020] [Accepted: 12/27/2020] [Indexed: 11/22/2022] Open
Abstract
Neutrophils release neutrophil extracellular traps (NET) comprising of decondensed chromatin that immobilizes and kills pathogens. In vitro generation of neutrophils on a large scale from hematopoietic stem cells (HSCs) may be a useful strategy for treating neutropenic patients in future, though it is not in clinical practice yet. Microbial infections lead to major cause of morbidity and mortality in these patients. Despite the importance of NET in preventing infection, efficacy of in vitro-generated neutrophils from HSCs to form NET is not tested. We show that functional neutrophils could be generated in vitro from HSCs/MNCs isolated from umbilical cord blood (UCB) and apheresis-derived peripheral blood (APBL). Neutrophils generated from UCB showed properties comparable to those isolated from peripheral blood. We also show that isolation of HSCs is not absolutely essential for in vitro neutrophil generation. Further, we show that neutrophils generated from HSCs express PADI4 enzyme and their NET-forming ability is comparable to peripheral blood neutrophils. Taken together, our data show that fully functional neutrophils can be generated in vitro from HSCs. NET-forming ability of in vitro-generated neutrophils is an important parameter to determine their functionality and thus, should be studied along with other standard functional assays.
Collapse
Affiliation(s)
- Rutuja Kuhikar
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India
| | - Nikhat Khan
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India
| | - Satyajeet P Khare
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University), Lavale, Pune 412115, India
| | - Amit Fulzele
- Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093, USA
| | - Sameer Melinkeri
- Blood and Marrow Transplant Unit, Deenanath Mangeshkar Hospital, Erandawne, Pune 411004, India
| | - Vaijayanti Kale
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India; Symbiosis Centre for Stem Cell Research, Symbiosis School of Biological Sciences, Symbiosis Knowledge Village, Lavale, Pune 412115, India
| | - Lalita Limaye
- National Centre for Cell Science, NCCS Complex, Savitribai Phule Pune University Campus, Pune 411007, India.
| |
Collapse
|
353
|
Abstract
Neutrophils produce neutrophil extracellular traps (NETs) by expelling their extracellular chromatin embedded with citrullinated histone H3, myeloperoxidase, and other intracellular molecules. Since their discovery in 2004, numerous articles have demonstrated the mechanism of NET formation and their function in innate immunity and inflammation. NET components often play an antimicrobial role, but excessive NETs are deleterious and can cause inflammation and tissue damage. This review highlights recent advancements in the identification of novel pathways and mechanisms of NET formation. We also focus on the specific damaging impact of NETs in individual organs. We then discuss the progress and limitations of various NET detection assays. Collectively, these vital aspects of NETs significantly improve our understanding of the pathobiology of NETs and future diagnostics and therapeutic tools for examining and modulating NETs in inflammatory diseases.
Collapse
Affiliation(s)
- Chuyi Tan
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Departments of Surgery and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
354
|
Teijeira A, Garasa S, Ochoa MC, Villalba M, Olivera I, Cirella A, Eguren-Santamaria I, Berraondo P, Schalper KA, de Andrea CE, Sanmamed MF, Melero I. IL8, Neutrophils, and NETs in a Collusion against Cancer Immunity and Immunotherapy. Clin Cancer Res 2020; 27:2383-2393. [PMID: 33376096 DOI: 10.1158/1078-0432.ccr-20-1319] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/18/2020] [Accepted: 12/23/2020] [Indexed: 12/12/2022]
Abstract
One of the most important mechanisms by which cancer fosters its own development is the generation of an immune microenvironment that inhibits or impairs antitumor immune responses. A cancer permissive immune microenvironment is present in a large proportion of the patients with cancer who do not respond to immunotherapy approaches intended to trigger preexisting antitumor immune responses, for instance, immune checkpoint blockade. High circulating levels of IL8 in patients with cancer quite accurately predict those who will not benefit from checkpoint-based immunotherapy. IL8 has been reported to favor cancer progression and metastases via different mechanisms, including proangiogenesis and the maintenance of cancer stem cells, but its ability to attract and functionally modulate neutrophils and macrophages is arguably one of the most important factors. IL8 does not only recruit neutrophils to tumor lesions, but also triggers the extrusion of neutrophil extracellular traps (NET). The relevance and mechanisms underlying the contribution of both neutrophils and NETs to cancer development and progression are starting to be uncovered and include both direct effects on cancer cells and changes in the tumor microenvironment, such as facilitating metastasis, awakening micrometastases from dormancy, and facilitating escape from cytotoxic immune cells. Blockade of IL8 or its receptors (CXCR1 and CXCR2) is being pursued in drug development, and clinical trials alone or in combination with anti-PD-L1 checkpoint inhibitors are already ongoing.
Collapse
Affiliation(s)
- Alvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Saray Garasa
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Maria C Ochoa
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Maria Villalba
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Irene Olivera
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Assunta Cirella
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Iñaki Eguren-Santamaria
- Departments of Oncology and Immunology, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Carlos E de Andrea
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Department of Pathology, Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Miguel F Sanmamed
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain.,Departments of Oncology and Immunology, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Navarra, Spain. .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Navarra Institute for Health Research (IDISNA), Pamplona, Navarra, Spain
| |
Collapse
|
355
|
Zhu Y, Denholtz M, Lu H, Murre C. Calcium signaling instructs NIPBL recruitment at active enhancers and promoters via distinct mechanisms to reconstruct genome compartmentalization. Genes Dev 2020; 35:65-81. [PMID: 33334824 PMCID: PMC7778268 DOI: 10.1101/gad.343475.120] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 10/22/2020] [Indexed: 12/18/2022]
Abstract
In this study, Zhu et al. sought to understand the signaling pathways and epigenetic control of nuclear architecture during developmental progression of immune cells. They found that in activated neutrophils calcium influx rapidly recruited the cohesin-loading factor NIPBL to thousands of active enhancers and promoters to dictate widespread changes in compartment segregation. During developmental progression the genomes of immune cells undergo large-scale changes in chromatin folding. However, insights into signaling pathways and epigenetic control of nuclear architecture remain rudimentary. Here, we found that in activated neutrophils calcium influx rapidly recruited the cohesin-loading factor NIPBL to thousands of active enhancers and promoters to dictate widespread changes in compartment segregation. NIPBL recruitment to enhancers and promoters occurred with distinct kinetics. The induction of NIPBL-binding was coordinate with increased P300, BRG1 and RNA polymerase II occupancy. NIPBL-bound enhancers were associated with NFAT, PU.1, and CEBP cis elements, whereas NIPBL-bound promoters were enriched for GC-rich DNA sequences. Using an acute degradation system, we found that the histone acetyltransferases P300 and CBP maintained H3K27ac abundance and facilitated NIPBL occupancy at enhancers and that active transcriptional elongation is essential to maintain H3K27ac abundance. Chromatin remodelers, containing either of the mutually exclusive BRG1 and BRM ATPases, promoted NIPBL recruitment at active enhancers. Conversely, at active promoters, depletion of BRG1 and BRM showed minimal effect on NIPBL occupancy. Finally, we found that calcium signaling in both primary innate and adaptive immune cells swiftly induced NIPBL occupancy. Collectively, these data reveal how transcriptional regulators, histone acetyltransferases, chromatin remodelers, and transcription elongation promote NIPBL occupancy at active enhancers while the induction of NIPLB occupancy at promoters is primarily associated with GC-rich DNA sequences.
Collapse
Affiliation(s)
- Yina Zhu
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Matthew Denholtz
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Hanbin Lu
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Cornelis Murre
- Division of Biological Sciences, Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
356
|
Lelliott PM, Momota M, Shibahara T, Lee MSJ, Smith NI, Ishii KJ, Coban C. Heparin induces neutrophil elastase-dependent vital and lytic NET formation. Int Immunol 2020; 32:359-368. [PMID: 31879779 DOI: 10.1093/intimm/dxz084] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/23/2019] [Indexed: 12/24/2022] Open
Abstract
Heparin is used extensively as an anticoagulant in a broad range of diseases and procedures; however, its biological effects are not limited to coagulation and remain incompletely understood. Heparin usage can lead to the life-threatening complication known as heparin-induced thrombocytopenia (HIT), caused by the development of antibodies against heparin/PF4 complexes. Here, we demonstrate the ability of heparin to induce neutrophil extracellular traps (NETs). NETs occurred with cell lysis and death, but live neutrophils releasing extracellular DNA strands, known as vital NETs, also occurred abundantly. Formation of NETs was time and dose dependent, and required reactive oxygen species and neutrophil elastase. Other compounds related to heparin such as low molecular weight heparin, fondaparinux and heparan sulfate either failed to induce NETs, or did so to a much lesser extent. Our findings suggest the ability of heparin to directly induce NET formation should be considered in the context of heparin treatment and HIT pathogenesis.
Collapse
Affiliation(s)
| | - Masatoshi Momota
- Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, Yamada-oka, Suita, Osaka, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Saitoasagi, Ibaraki, Osaka, Japan
| | - Takayuki Shibahara
- Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, Yamada-oka, Suita, Osaka, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Saitoasagi, Ibaraki, Osaka, Japan
| | - Michelle S J Lee
- Laboratory of Malaria Immunology, Yamada-oka, Suita, Osaka, Japan
| | - Nicholas I Smith
- Biophotonics Laboratory, Immunology Frontier Research Center (IFReC), Osaka University, Yamada-oka, Suita, Osaka, Japan
| | - Ken J Ishii
- Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, Yamada-oka, Suita, Osaka, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Saitoasagi, Ibaraki, Osaka, Japan.,Division of Vaccine Science, Shirokanedai, Minato-ku, Tokyo, Japan
| | - Cevayir Coban
- Laboratory of Malaria Immunology, Yamada-oka, Suita, Osaka, Japan.,Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639 Tokyo, Japan
| |
Collapse
|
357
|
Vorobjeva NV, Chernyak BV. NETosis: Molecular Mechanisms, Role in Physiology and Pathology. BIOCHEMISTRY (MOSCOW) 2020; 85:1178-1190. [PMID: 33202203 PMCID: PMC7590568 DOI: 10.1134/s0006297920100065] [Citation(s) in RCA: 258] [Impact Index Per Article: 51.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
NETosis is a program for formation of neutrophil extracellular traps (NETs), which consist of modified chromatin decorated with bactericidal proteins from granules and cytoplasm. Various pathogens, antibodies and immune complexes, cytokines, microcrystals, and other physiological stimuli can cause NETosis. Induction of NETosis depends on reactive oxygen species (ROS), the main source of which is NADPH oxidase. Activation of NADPH oxidase depends on increase in the concentration of Ca2+ in the cytoplasm and in some cases on the generation of ROS in mitochondria. NETosis includes release of the granule components into the cytosol, modification of histones leading to chromatin decondensation, destruction of the nuclear envelope, as well as formation of pores in the plasma membrane. In this review, basic mechanisms of NETosis, as well as its role in the pathogenesis of some diseases including COVID-19 are discussed.
Collapse
Affiliation(s)
- N V Vorobjeva
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia.
| | - B V Chernyak
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119992, Russia.
| |
Collapse
|
358
|
Williams TL, Rada B, Tandon E, Gestal MC. "NETs and EETs, a Whole Web of Mess". Microorganisms 2020; 8:E1925. [PMID: 33291570 PMCID: PMC7761834 DOI: 10.3390/microorganisms8121925] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Neutrophils and eosinophils are granulocytes that have very distinct functions. Neutrophils are first responders to external threats, and they use different mechanisms to control pathogens. Phagocytosis, reactive oxygen species, and neutrophil extracellular traps (NETs) are some of the mechanisms that neutrophils utilize to fight pathogens. Although there is some controversy as to whether NETs are in fact beneficial or detrimental to the host, it mainly depends on the biological context. NETs can contribute to disease pathogenesis in certain types of diseases, while they are also undeniably critical components of the innate immune response. On the contrary, the role of eosinophils during host immune responses remains to be better elucidated. Eosinophils play an important role during helminthic infections and allergic responses. Eosinophils can function as effector cells in viral respiratory infections, gut bacterial infections, and as modulators of immune responses by driving the balance between Th1 and Th2 responses. In particular, eosinophils have biological activities that appear to be quite similar to those of neutrophils. Both possess bactericidal activity, can activate proinflammatory responses, can modulate adaptive immune responses, can form extracellular traps, and can be beneficial or detrimental to the host according to the underlying pathology. In this review we compare these two cell types with a focus on highlighting their numerous similarities related to extracellular traps.
Collapse
Affiliation(s)
- Tyler L. Williams
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| | - Balázs Rada
- Department of Infectious Diseases, University of Georgia, Athens, GA 30302, USA;
| | - Eshaan Tandon
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| | - Monica C. Gestal
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| |
Collapse
|
359
|
Yang J, Wu Z, Long Q, Huang J, Hong T, Liu W, Lin J. Insights Into Immunothrombosis: The Interplay Among Neutrophil Extracellular Trap, von Willebrand Factor, and ADAMTS13. Front Immunol 2020; 11:610696. [PMID: 33343584 PMCID: PMC7738460 DOI: 10.3389/fimmu.2020.610696] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 11/02/2020] [Indexed: 12/24/2022] Open
Abstract
Both neutrophil extracellular traps (NETs) and von Willebrand factor (VWF) are essential for thrombosis and inflammation. During these processes, a complex series of events, including endothelial activation, NET formation, VWF secretion, and blood cell adhesion, aggregation and activation, occurs in an ordered manner in the vasculature. The adhesive activity of VWF multimers is regulated by a specific metalloprotease ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motifs, member 13). Increasing evidence indicates that the interaction between NETs and VWF contributes to arterial and venous thrombosis as well as inflammation. Furthermore, contents released from activated neutrophils or NETs induce the reduction of ADAMTS13 activity, which may occur in both thrombotic microangiopathies (TMAs) and acute ischemic stroke (AIS). Recently, NET is considered as a driver of endothelial damage and immunothrombosis in COVID-19. In addition, the levels of VWF and ADAMTS13 can predict the mortality of COVID-19. In this review, we summarize the biological characteristics and interactions of NETs, VWF, and ADAMTS13, and discuss their roles in TMAs, AIS, and COVID-19. Targeting the NET-VWF axis may be a novel therapeutic strategy for inflammation-associated TMAs, AIS, and COVID-19.
Collapse
Affiliation(s)
- Junxian Yang
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Zhiwei Wu
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Quan Long
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Jiaqi Huang
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Tiantian Hong
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Wang Liu
- Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Jiangguo Lin
- Research Department of Medical Sciences, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,Institute of Biomechanics/School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| |
Collapse
|
360
|
Imre G. Cell death signalling in virus infection. Cell Signal 2020; 76:109772. [PMID: 32931899 PMCID: PMC7486881 DOI: 10.1016/j.cellsig.2020.109772] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Apoptosis, necroptosis and pyroptosis represent three major regulated cell death modalities. Apoptosis features cell shrinkage, nuclear fragmentation and cytoplasm-blebbing. Necroptosis and pyroptosis exhibit osmotic imbalances in the cell accompanied by early membrane ruptures, which morphologically resembles necrosis. Importantly, these two lytic cell death forms facilitate the release of damage associated molecular patterns into the extracellular space leading to inflammatory response. Whereas, during apoptosis, the membrane integrity is preserved and the apoptotic cell is removed by neighbouring cells ensuring the avoidance of immune-stimulation. Viruses comprise a versatile group of intracellular pathogens, which elicit various strategies to infect and to propagate. Viruses are recognized by a myriad of pathogen recognition receptors in the human cells, which consequently lead to activation of the immune system and in certain circumstances cell-autonomous cell death. Importantly, the long-standing view that a cell death inducing capacity of a virus is equal to its pathogenic potential seems to be only partially valid. The altruistic cell death of an infected cell may serve the whole organism by ultimately curbing the way of virus manufacturing. In fact, several viruses express "anti-cell death" proteins to avoid this viral-defence mechanism. Conversely, some viruses hijack cell death pathways to selectively destroy cell populations in order to compromise the immune system of the host. This review discusses the pros and cons of virus induced cell death from the perspective of the host cells and attempts to provide a comprehensive overview of the complex network of cell death signalling in virus infection.
Collapse
Affiliation(s)
- Gergely Imre
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.
| |
Collapse
|
361
|
Urban CF, Backman E. Eradicating, retaining, balancing, swarming, shuttling and dumping: a myriad of tasks for neutrophils during fungal infection. Curr Opin Microbiol 2020; 58:106-115. [DOI: 10.1016/j.mib.2020.09.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/26/2022]
|
362
|
Mazzoleni V, Zimmermann K, Smirnova A, Tarassov I, Prévost G. Staphylococcus aureus Panton-Valentine Leukocidin triggers an alternative NETosis process targeting mitochondria. FASEB J 2020; 35:e21167. [PMID: 33241563 DOI: 10.1096/fj.201902981r] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 09/18/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023]
Abstract
Panton-Valentine Leukocidin (PVL) is a bicomponent leukotoxin produced by 3%-10% of clinical Staphylococcus aureus (SA) strains involved in the severity of hospital and community-acquired infections. Although PVL was long known as a pore-forming toxin, recent studies have challenged the formation of a pore at the plasma membrane, while its endocytosis and the exact mode of action remain to be defined. In vitro immunolabeling of human neutrophils shows that Neutrophil Extracellular Traps (NETosis) is triggered by the action of purified PVL, but not by Gamma hemolysin CB (HlgCB), a structurally similar SA leukotoxin. PVL causes the ejection of chromatin fibers (NETs) decorated with antibacterial peptides independently of the NADPH oxidase oxidative burst. Leukotoxin partially colocalizes with mitochondria and enhances the production of reactive oxygen species from these organelles, while showing an increased autophagy, which results unnecessary for NETs ejection. PVL NETosis is elicited through Ca2+ -activated SK channels and Myeloperoxidase activity but is abolished by Allopurinol pretreatment of neutrophils. Moreover, massive citrullination of the histone H3 is performed by peptidyl arginine deiminases. Inhibition of this latter enzymes fails to abolish NET extrusion. Unexpectedly, PVL NETosis does not seem to involve Src kinases, which is the main kinase family activated downstream the binding of PVL F subunit to CD45 receptor, while the specific kinase pathway differs from the NADPH oxidase-dependent NETosis. PVL alone causes a different and specific form of NETosis that may rather represent a bacterial strategy conceived to disarm and disrupt the immune response, eventually allowing SA to spread.
Collapse
Affiliation(s)
- Viola Mazzoleni
- University of Strasbourg, CHRU Strasbourg, ITI InnoVec, Fédération de Médecine Translationnelle de Strasbourg, UR7290, Institut de Bactériologie, Strasbourg, France
| | - Kiran Zimmermann
- University of Strasbourg, CHRU Strasbourg, ITI InnoVec, Fédération de Médecine Translationnelle de Strasbourg, UR7290, Institut de Bactériologie, Strasbourg, France
| | - Anna Smirnova
- UMR 7156 GMGM Strasbourg University/CNRS, Strasbourg, France
| | - Ivan Tarassov
- UMR 7156 GMGM Strasbourg University/CNRS, Strasbourg, France
| | - Gilles Prévost
- University of Strasbourg, CHRU Strasbourg, ITI InnoVec, Fédération de Médecine Translationnelle de Strasbourg, UR7290, Institut de Bactériologie, Strasbourg, France
| |
Collapse
|
363
|
Hu J, Kang H, Chen H, Yao J, Yi X, Tang W, Wan M. Targeting neutrophil extracellular traps in severe acute pancreatitis treatment. Therap Adv Gastroenterol 2020; 13:1756284820974913. [PMID: 33281940 PMCID: PMC7692350 DOI: 10.1177/1756284820974913] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/26/2020] [Indexed: 02/05/2023] Open
Abstract
Severe acute pancreatitis (SAP) is a critical abdominal disease associated with high death rates. A systemic inflammatory response promotes disease progression, resulting in multiple organ dysfunction. The functions of neutrophils in the pathology of SAP have been presumed traditionally to be activation of chemokine and cytokine cascades accompanying the inflammatory process. Recently, since their discovery, a new type of antimicrobial mechanism, neutrophil extracellular traps (NETs), and their role in SAP, has attracted widespread attention from the scientific community. Significantly different from phagocytosis and degranulation, NETs kill extracellular microorganisms by releasing DNA fibers decorated with granular proteins. In addition to their strong antimicrobial functions, NETs participate in the pathophysiological process of many noninfectious diseases. In SAP, NETs injure normal tissues under inflammatory stress, which is associated with the activation of inflammatory cells, to cause an inflammatory cascade, and SAP products also trigger NET formation. Thus, due to the interaction between NET generation and SAP, a treatment targeting NETs might become a key point in SAP therapy. In this review, we summarize the mechanism of NETs in protecting the host from pathogen invasion, the stimulus that triggers NET formation, organ injury associated with SAP involving NETs, methods to interrupt the harmful effects of NETs, and different therapeutic strategies to preserve the organ function of patients with SAP by targeting NETs.
Collapse
Affiliation(s)
| | | | - Huan Chen
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jiaqi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaolin Yi
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wenfu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
364
|
Krogh AKH, Lyngby JG, Bjørnvad CR, Nielsen LN. Presence of nucleosomes in plasma and increased thrombin generation in dogs with acute and chronic gastroenteropathies. Res Vet Sci 2020; 135:504-510. [PMID: 33243453 DOI: 10.1016/j.rvsc.2020.11.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023]
Abstract
Neutrophil extracellular traps (NETs) which contain nucleosomes protect the host by eliminating extracellular pathogens. However, any inflammatory stimuli can activate NETs and eventually lead to an immune overreaction leading to autoimmune diseases and thrombosis. Acute/chronic gastroenteropathies(aGE/cGE) are prevalent in dogs, and are associated with a strong inflammatory component. The aim of this study was to investigate if dogs with aGE and cGE have increased concentrations of nucleosomes indicative of NETs formation, and whether increased concentrations of nucleosomes are associated with hypercoagulability determined by increased thrombin generation. Twenty-six dogs were enrolled. The dogs were healthy (n = 11), or presented with aGE(n = 7) or cGE(n = 8). Minimum database including CRP, APTT, PT and fibrinogen, was obtained from all dogs. Citrated plasma was batched and used for subsequent analyses. Nucleosome concentration was analysed using a Cell-Death Detection ELISA-kit and thrombin generation by a calibrated automated thrombogram assay. No statistical differences in nucleosome concentrations were present between the groups. Although a numerically increased concentration of nucleosomes where seen in dogs with aGE(median;range) (0.019 AU;0.003-0.088) and cGE(0.023 AU;0.011-0.256) compared to controls(0.007 AU;0.003-0.042). One dog with GI-lymphoma demonstrated a markedly increased concentration of nucleosomes (0.256 AU). Dogs with aGE showed increased thrombin generation by increased peak (p = 0.03) and endogenous thrombin potential (p = 0.03); and increased CRP (p = 0.001), fibrinogen (p = 0.0002) and prolonged APTT (p = 0.03) compared to controls. This proof of concept study demonstrates that dogs with aGE and cGE have presence of nucleosomes with marked increase in one dog with GI-lymphoma. Nucleosomes might be linked to haemostatic alterations in dogs with inflammatory and neoplastic diseases.
Collapse
Affiliation(s)
- A K H Krogh
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark.
| | - J G Lyngby
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - C R Bjørnvad
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - L N Nielsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
365
|
Li M, Lin C, Leso A, Nefedova Y. Quantification of Citrullinated Histone H3 Bound DNA for Detection of Neutrophil Extracellular Traps. Cancers (Basel) 2020; 12:cancers12113424. [PMID: 33218159 PMCID: PMC7698949 DOI: 10.3390/cancers12113424] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/15/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
Formation of neutrophil extracellular traps (NETs) has been associated with multiple pathologies including cancer. While the visualization of NETs by microscopy is a routine technique, their quantification presents a number of challenges. Commonly, as citrullination of histone H3 is required for NET formation, the presence of this modified histone along with DNA is considered to be a hallmark of NETs. Here, we describe and validate a novel assay for the quantification of NETs based on the detection of citrullinated histone H3 bound to DNA (CitH3DNA binding assay). Using this assay, we investigated the effect of phorbol 12-myristate 13-acetate (PMA) on NET formation by neutrophils isolated from the bone marrow of control and myeloma-bearing mice. We found that PMA induced citrullination of histone H3, an increase in the level of CitH3DNA, and NET formation in neutrophils from both tumor-free and myeloma-bearing mice. The levels of CitH3DNA in the NET fractions, as measured by our assay, directly correlated with the citrullination of histone H3 in neutrophils, as detected by Western blotting, and were significantly higher in PMA-stimulated compared to unstimulated neutrophils. Neutrophils from tumor-bearing mice produced more NETs than those from tumor-free counterparts following stimulation with PMA. The increase in NET production correlated with significantly higher histone H3 citrullination levels and increased measurements of CitH3DNA. Thus, our data indicate that bone marrow neutrophils from myeloma-bearing hosts are prone to NET formation.
Collapse
|
366
|
Alhede M, Alhede M, Qvortrup K, Kragh KN, Jensen PØ, Stewart PS, Bjarnsholt T. The origin of extracellular DNA in bacterial biofilm infections in vivo. Pathog Dis 2020; 78:5810662. [PMID: 32196074 PMCID: PMC7150582 DOI: 10.1093/femspd/ftaa018] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/19/2020] [Indexed: 02/03/2023] Open
Abstract
Extracellular DNA (eDNA) plays an important role in both the aggregation of bacteria and in the interaction of the resulting biofilms with polymorphonuclear leukocytes (PMNs) during an inflammatory response. Here, transmission electron and confocal scanning laser microscopy were used to examine the interaction between biofilms of Pseudomonas aeruginosa and PMNs in a murine implant model and in lung tissue from chronically infected cystic fibrosis patients. PNA FISH, DNA staining, labeling of PMN DNA with a thymidine analogue and immunohistochemistry were applied to localize bacteria, eDNA, PMN-derived eDNA, PMN-derived histone H3 (H3), neutrophil elastase (NE) and citrullinated H3 (citH3). Host-derived eDNA was observed surrounding bacterial biofilms but not within the biofilms. H3 localized to the lining of biofilms while NE was found throughout biofilms. CitH3, a marker for neutrophil extracellular traps (NETs) was detected only sporadically indicating that most host-derived eDNA in vivo was not a result of NETosis. Together these observations show that, in these in vivo biofilm infections with P. aeruginosa, the majority of eDNA is found external to the biofilm and derives from the host.
Collapse
Affiliation(s)
- Maria Alhede
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark
| | - Morten Alhede
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark
| | - Klaus Qvortrup
- CFIM/Department of Biomedical Sciences, University of Copenhagen, Blegdmasvej 3, DK-2200 Copenhagen N., Denmark
| | - Kasper Nørskov Kragh
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark
| | - Peter Østrup Jensen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark.,Department of Clinical Microbiology, H:S Rigshospitalet, Juliane Maries Vej 22, DK-2100 Copenhagen Ø., Denmark
| | - Philip Shook Stewart
- Center for Biofilm Engineering, Montana State University, 366 Barnard Hall, P.O. Box 173980, Bozeman, MT 59717-3980, USA
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark.,Department of Clinical Microbiology, H:S Rigshospitalet, Juliane Maries Vej 22, DK-2100 Copenhagen Ø., Denmark
| |
Collapse
|
367
|
Haritha VH, George A, Shaji BV, Anie Y. NET-associated citrullinated histones promote LDL aggregation and foam cell formation in vitro. Exp Cell Res 2020; 396:112320. [PMID: 33058833 DOI: 10.1016/j.yexcr.2020.112320] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 11/16/2022]
Abstract
Neutrophils have been recently identified in the atherosclerotic lesion and they can release neutrophil extracellular trap (NET) under the pro-inflammatory conditions prevailing in the lesion. Citrullinated histones (Cit-histones) are the major type of citrullinated proteins associated with NET release. Since elevated levels of citrullinated proteins have been detected in inflammatory diseases including atherosclerosis, this study analysed the role played by NET and Cit-histones in different atherogenic events in vitro. First, neutrophil recruitment and NET release in the presence of low-density lipoprotein (LDL) and oxidised LDL (Ox-LDL) were analysed by Boyden's chamber method and microscopy respectively. Then, LDL oxidation and LDL aggregation in the presence of NET and Cit-histones were analysed spectroscopically. Foam cell formation in the presence of NET or Cit-histone was studied by both microscopic and spectroscopic methods. While neutrophil recruitment was facilitated by Ox-LDL and not by LDL, the extent of NET release was significantly increased in the presence of both LDL and Ox-LDL. In the presence of NET, LDL oxidation, aggregation and foam cell formation were found to be increased. Cit-histones were found to accelerate LDL aggregation and foam cell formation at higher citrulline levels. Altogether, the results suggest that both NET and NET-associated Cit-histone released at the lesion can play major roles as pro-atherogenic mediators. Inhibiting the action of NET or Cit-histone would, therefore, be beneficial in slowing down atherosclerotic progression.
Collapse
Affiliation(s)
- V H Haritha
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India.
| | - Anjana George
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India.
| | - Binchu V Shaji
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India.
| | - Y Anie
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India.
| |
Collapse
|
368
|
Jia J, Wang M, Ma Y, Teng J, Shi H, Liu H, Sun Y, Su Y, Meng J, Chi H, Chen X, Cheng X, Ye J, Liu T, Wang Z, Wan L, Zhou Z, Wang F, Yang C, Hu Q. Circulating Neutrophil Extracellular Traps Signature for Identifying Organ Involvement and Response to Glucocorticoid in Adult-Onset Still's Disease: A Machine Learning Study. Front Immunol 2020; 11:563335. [PMID: 33240258 PMCID: PMC7680913 DOI: 10.3389/fimmu.2020.563335] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
Adult-onset Still’s disease (AOSD) is an autoinflammatory disease with multisystem involvement. Early identification of patients with severe complications and those refractory to glucocorticoid is crucial to improve therapeutic strategy in AOSD. Exaggerated neutrophil activation and enhanced formation of neutrophil extracellular traps (NETs) in patients with AOSD were found to be closely associated with etiopathogenesis. In this study, we aim to investigate, to our knowledge for the first time, the clinical value of circulating NETs by machine learning to distinguish AOSD patients with organ involvement and refractory to glucocorticoid. Plasma samples were used to measure cell-free DNA, NE-DNA, MPO-DNA, and citH3-DNA complexes from training and validation sets. The training set included 40 AOSD patients and 24 healthy controls (HCs), and the validation set included 26 AOSD patients and 16 HCs. Support vector machines (SVM) were used for modeling and validation of circulating NETs signature for the diagnosis of AOSD and identifying patients refractory to low-dose glucocorticoid treatment. The training set was used to build a model, and the validation set was used to test the predictive capacity of the model. A total of four circulating NETs showed similar trends in different individuals and could distinguish patients with AOSD from HCs by SVM (AUC value: 0.88). Circulating NETs in plasma were closely correlated with systemic score, laboratory tests, and cytokines. Moreover, circulating NETs had the potential to distinguish patients with liver and cardiopulmonary system involvement. Furthermore, the AUC value of combined NETs to identify patients who were refractory to low-dose glucocorticoid was 0.917. In conclusion, circulating NETs signature provide added clinical value in monitoring AOSD patients. It may provide evidence to predict who is prone to be refractory to low-dose glucocorticoid and help to make efficient therapeutic strategy.
Collapse
Affiliation(s)
- Jinchao Jia
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mengyan Wang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuning Ma
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialin Teng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Shi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Honglei Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Sun
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yutong Su
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianfen Meng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Rheumatology and Immunology, The First People's Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| | - Huihui Chi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Chen
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaobing Cheng
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junna Ye
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihong Wang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liyan Wan
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuochao Zhou
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Wang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengde Yang
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiongyi Hu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
369
|
Ríos-López AL, González GM, Hernández-Bello R, Sánchez-González A. Avoiding the trap: Mechanisms developed by pathogens to escape neutrophil extracellular traps. Microbiol Res 2020; 243:126644. [PMID: 33199088 DOI: 10.1016/j.micres.2020.126644] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022]
Abstract
Neutrophils are the first cells of the innate immune system that respond to infection by arriving at sites when pathogens have exceeded physical barriers. Among their response mechanisms against pathogens is the release of neutrophil extracellular traps (NETs), which are composed of deoxyribonucleic acid and antimicrobial proteins such as neutrophil elastase, myeloperoxidase, antimicrobial peptides, and other proteins in neutrophil granules. The formation of extracellular traps is considered an effective strategy to capture and, in some cases, neutralize pathogenic bacteria, fungi, parasites, or viruses. However, it is also known that pathogens can respond to NETs by expressing some virulence factors, thus evading the antimicrobial effect of these structures. These include the secretion of proteins to degrade the deoxyribonucleic acid scaffold, the formation of biofilms that impede the effect of NETs, or the modification of its membrane structure to avoid interaction with NETs. In this review, we discuss these mechanisms and summarize the different pathogens that employ one or more mechanisms to evade the NET-mediated neutrophil response.
Collapse
Affiliation(s)
- A L Ríos-López
- Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, 64460, Mexico
| | - G M González
- Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, 64460, Mexico
| | - R Hernández-Bello
- Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, 64460, Mexico
| | - A Sánchez-González
- Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, 64460, Mexico.
| |
Collapse
|
370
|
Putative Origins of Cell-Free DNA in Humans: A Review of Active and Passive Nucleic Acid Release Mechanisms. Int J Mol Sci 2020; 21:ijms21218062. [PMID: 33137955 PMCID: PMC7662960 DOI: 10.3390/ijms21218062] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Through various pathways of cell death, degradation, and regulated extrusion, partial or complete genomes of various origins (e.g., host cells, fetal cells, and infiltrating viruses and microbes) are continuously shed into human body fluids in the form of segmented cell-free DNA (cfDNA) molecules. While the genetic complexity of total cfDNA is vast, the development of progressively efficient extraction, high-throughput sequencing, characterization via bioinformatics procedures, and detection have resulted in increasingly accurate partitioning and profiling of cfDNA subtypes. Not surprisingly, cfDNA analysis is emerging as a powerful clinical tool in many branches of medicine. In addition, the low invasiveness of longitudinal cfDNA sampling provides unprecedented access to study temporal genomic changes in a variety of contexts. However, the genetic diversity of cfDNA is also a great source of ambiguity and poses significant experimental and analytical challenges. For example, the cfDNA population in the bloodstream is heterogeneous and also fluctuates dynamically, differs between individuals, and exhibits numerous overlapping features despite often originating from different sources and processes. Therefore, a deeper understanding of the determining variables that impact the properties of cfDNA is crucial, however, thus far, is largely lacking. In this work we review recent and historical research on active vs. passive release mechanisms and estimate the significance and extent of their contribution to the composition of cfDNA.
Collapse
|
371
|
Rosazza T, Warner J, Sollberger G. NET formation - mechanisms and how they relate to other cell death pathways. FEBS J 2020; 288:3334-3350. [PMID: 33047496 DOI: 10.1111/febs.15589] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/29/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022]
Abstract
Cell death is an integral part of both infectious and sterile inflammatory reactions. Many cell death pathways cause the dying cell to lyse, thereby amplifying inflammation. A special form of lytic cell death is the formation of neutrophil extracellular traps (NETs), large structures of chromatin and antimicrobial proteins, which are released by dying neutrophils to capture extracellular pathogens and limit the spread of infections. The molecular mechanisms of NET formation remain incompletely understood. Recent research demonstrated substantial crosstalk between different cell death pathways, most notably between apoptosis, pyroptosis and necroptosis. Here, we review suicidal and vital NET formation and discuss potential crosstalk of their mechanisms of release with other forms of cell death.
Collapse
Affiliation(s)
- Thibault Rosazza
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, UK
| | - Jordan Warner
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, UK
| | - Gabriel Sollberger
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, UK
| |
Collapse
|
372
|
Macedo IS, Lima MVA, Souza JS, Rochael NC, Caldas PN, Barbosa HS, Lara FA, Saraiva EM, Mariante RM. Extracellular Traps Released by Neutrophils from Cats are Detrimental to Toxoplasma gondii Infectivity. Microorganisms 2020; 8:microorganisms8111628. [PMID: 33105542 PMCID: PMC7716220 DOI: 10.3390/microorganisms8111628] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/03/2020] [Accepted: 10/11/2020] [Indexed: 12/18/2022] Open
Abstract
Toxoplasma gondii is the causative agent of toxoplasmosis, an infectious disease that affects over 30% of the human world population, causing fatal infections in immunocompromised individuals and neonates. The life cycle of T. gondii is complex, and involves intermediate hosts (birds and mammals) and definitive hosts (felines, including domestic cats). The innate immune repertoire against the parasite involves the production of neutrophil extracellular traps (NET), and neutrophils from several intermediate hosts produce NET induced by T. gondii. However, the mechanisms underlying NET release in response to the parasite have been poorly explored. Therefore, the aims of this study were to investigate whether neutrophils from cats produce NET triggered by T. gondii and to understand the mechanisms thereby involved. Neutrophils from cats were stimulated with T. gondii tachyzoites and NET-derived DNA in the supernatant was quantified during the time. The presence of histone H1 and myeloperoxidase was detected by immunofluorescence. We observed that cat neutrophils produce both classical and rapid/early NET stimulated by T. gondii. Inhibition of elastase, intracellular calcium, and phosphatidylinositol 3-kinase (PI3K)-δ partially blocked classical NET release in response to the parasite. Electron microscopy revealed strands and networks of DNA in close contact or completely entrapping parasites. Live imaging showed that tachyzoites are killed by NET. We conclude that the production of NET is a conserved strategy to control infection by T. gondii amongst intermediate and definitive hosts.
Collapse
Affiliation(s)
- Isabela S. Macedo
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (I.S.M.); (M.V.A.L.); (J.S.S.); (H.S.B.)
| | - Marcos V. A. Lima
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (I.S.M.); (M.V.A.L.); (J.S.S.); (H.S.B.)
| | - Jéssica S. Souza
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (I.S.M.); (M.V.A.L.); (J.S.S.); (H.S.B.)
| | - Natalia C. Rochael
- Laboratório de Imunobiologia das Leishmanioses, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (N.C.R.); (E.M.S.)
| | - Pedro N. Caldas
- HVN Hospital Veterinário Niterói, Niterói 24360-440, RJ, Brazil;
| | - Helene S. Barbosa
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (I.S.M.); (M.V.A.L.); (J.S.S.); (H.S.B.)
| | - Flávio A. Lara
- Laboratório de Microbiologia Celular, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil;
| | - Elvira M. Saraiva
- Laboratório de Imunobiologia das Leishmanioses, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil; (N.C.R.); (E.M.S.)
| | - Rafael M. Mariante
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, RJ, Brazil; (I.S.M.); (M.V.A.L.); (J.S.S.); (H.S.B.)
- Correspondence: or ; Tel.: +55-21-2562-1018
| |
Collapse
|
373
|
Edwards NJ, Hwang C, Marini S, Pagani CA, Spreadborough PJ, Rowe CJ, Yu P, Mei A, Visser N, Li S, Hespe GE, Huber AK, Strong AL, Shelef MA, Knight JS, Davis TA, Levi B. The role of neutrophil extracellular traps and TLR signaling in skeletal muscle ischemia reperfusion injury. FASEB J 2020; 34:15753-15770. [PMID: 33089917 DOI: 10.1096/fj.202000994rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 08/20/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
Ischemia reperfusion (IR) injury results in devastating skeletal muscle fibrosis. Here, we recapitulate this injury with a mouse model of hindlimb IR injury which leads to skeletal muscle fibrosis. Injury resulted in extensive immune infiltration with robust neutrophil extracellular trap (NET) formation in the skeletal muscle, however, direct targeting of NETs via the peptidylarginine deiminase 4 (PAD4) mechanism was insufficient to reduce muscle fibrosis. Circulating levels of IL-10 and TNFα were significantly elevated post injury, indicating toll-like receptor (TLR) signaling may be involved in muscle injury. Administration of hydroxychloroquine (HCQ), a small molecule inhibitor of TLR7/8/9, following injury reduced NET formation, IL-10, and TNFα levels and ultimately mitigated muscle fibrosis and improved myofiber regeneration following IR injury. HCQ treatment decreased fibroadipogenic progenitor cell proliferation and partially inhibited ERK1/2 phosphorylation in the injured tissue, suggesting it may act through a combination of TLR7/8/9 and ERK signaling mechanisms. We demonstrate that treatment with FDA-approved HCQ leads to decreased muscle fibrosis and increased myofiber regeneration following IR injury, suggesting short-term HCQ treatment may be a viable treatment to prevent muscle fibrosis in ischemia reperfusion and traumatic extremity injury.
Collapse
Affiliation(s)
- Nicole J Edwards
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Charles Hwang
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Simone Marini
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Chase A Pagani
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Philip J Spreadborough
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Cassie J Rowe
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Pauline Yu
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Annie Mei
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Noelle Visser
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Shuli Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Geoffrey E Hespe
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Amanda K Huber
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Amy L Strong
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Miriam A Shelef
- Division of Rheumatology, University of Wisconsin and William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Jason S Knight
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Thomas A Davis
- Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.,Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
374
|
da Silva RF, Baptista D, Roth A, Miteva K, Burger F, Vuilleumier N, Carbone F, Montecucco F, Mach F, J. Brandt K. Anti-Apolipoprotein A-1 IgG Influences Neutrophil Extracellular Trap Content at Distinct Regions of Human Carotid Plaques. Int J Mol Sci 2020; 21:7721. [PMID: 33086507 PMCID: PMC7588926 DOI: 10.3390/ijms21207721] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neutrophils accumulate in atherosclerotic plaques. Neutrophil extracellular traps (NET) were recently identified in experimental atherosclerosis and in complex human lesions. However, not much is known about the NET marker citrullinated histone-3 (H3Cit) expression and functionality in human carotid plaques. Moreover, the association between the proatherosclerotic autoantibody anti-apolipoprotein A-1 (anti-ApoA-1 IgG) and NET has never been investigated. METHODS Atherosclerotic plaques have been obtained from 36 patients with severe carotid stenosis that underwent carotid endarterectomy for severe carotid stenosis. Samples were sectioned into upstream and downstream regions from the same artery segment. Plaque composition and expression of NET markers neutrophil elastase (NE) and H3Cit were quantified by immunohistochemistry. H3Cit expression and function was evaluated by immunofluorescence and confocal analysis in a subset of patients. RESULTS Pathological features of vulnerable phenotypes were exacerbated in plaques developed at downstream regions, including higher accumulation of neutrophils and enhanced expression of NE and H3Cit, as compared to plaques from upstream regions. The H3Cit signal was also more intense in downstream regions, with significant extracellular distribution in spaces outside of neutrophils. The percentage of H3Cit colocalization with CD66b (neutrophils) was markedly lower in downstream portions of carotid plaques, confirming the extrusion of NET in this region. In agreement, the maximum distance of the H3Cit signal from neutrophils, extrapolated from vortex distance calculation in all possible directions, was also higher in downstream plaques. The serum anti-ApoA-1index positively correlated with the expression of H3Cit in downstream segments of plaques. Expression of the H3Cit signal outside of neutrophils and H3Cit maximal distance from CD66b-positive cells increased in plaques from serum positive anti-ApoA-1 patients compared with serum negative patients. CONCLUSION NET elements are differentially expressed in upstream versus downstream regions of human carotid plaques and may be influenced by circulating levels of anti-ApoA-1 IgG. These findings could warrant the investigation of NET elements as potential markers of vulnerability.
Collapse
Affiliation(s)
- Rafaela F. da Silva
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Daniela Baptista
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Aline Roth
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Kapka Miteva
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Fabienne Burger
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Nicolas Vuilleumier
- Department of Diagnostics, Division of Laboratory Medicine, Geneva University Hospitals, 1211 Geneva, Switzerland;
- Department of Medical Specialities, Division of Laboratory Medicine, Faculty of Medicine, 1211 Geneva, Switzerland
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV n6, 16132 Genoa, Italy; (F.C.); (F.M.)
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Largo Rosanna Benzi n10, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV n6, 16132 Genoa, Italy; (F.C.); (F.M.)
- IRCCS Ospedale Policlinico San Martino Genoa-Italian Cardiovascular Network, Largo Rosanna Benzi n10, 16132 Genoa, Italy
| | - François Mach
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| | - Karim J. Brandt
- Division of Cardiology, Foundation for Medical Researches, Department of Medicine Specialties, Faculty of Medicine, University of Geneva, Av. de la Roseraie 64, 1211 Geneva, Switzerland; (R.F.d.S.); (D.B.); (A.R.); (K.M.); (F.B.); (F.M.)
| |
Collapse
|
375
|
Wang X, Blanco LP, Carmona-Rivera C, Nakabo S, Pedersen HL, Yu ZX, Kaplan MJ. Effects of Gasdermin D in Modulating Murine Lupus and its Associated Organ Damage. Arthritis Rheumatol 2020; 72:2118-2129. [PMID: 32692482 DOI: 10.1002/art.41444] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/02/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Gasdermin D (GSDMD) is the key executioner of an inflammatory cell death mechanism known as pyroptosis. Recent reports have also implicated GSDMD in other mechanisms of cell death, including apoptosis, necroptosis, and NETosis. Given the role of dysregulated cell death in autoimmune syndromes such as systemic lupus erythematosus (SLE), this study was undertaken in a murine lupus model to investigate whether GSDMD plays a pathogenic role in systemic autoimmunity by promoting inflammatory cell death, leading to increased generation of nuclear autoantigens and autoantibodies. METHODS An imiquimod-induced model of SLE was tested in GSDMD-/- mice (n = 30), with wild-type (WT) mice as controls (n = 34), on a C57BL/6 background. At the time of euthanasia, the mice were examined for serum autoantibodies, immune complex deposition, organ inflammation, immune dysregulation, and type I interferon responses. A model of pristane-induced lung injury in GSDMD-/- mice (n = 7), with WT mice as controls (n = 10), was used to confirm the pulmonary phenotype. Regulation of various mechanisms of cell death by GSDMD was investigated in the mice. RESULTS Unexpectedly, GSDMD-/- mice developed enhanced mortality, more severe renal and pulmonary inflammation, and exacerbated autoantibody production in response to imiquimod. Pulmonary involvement was also more severe in the absence of GSDMD in mice with pristane-induced lung injury. Compared to WT mice, lack of GSDMD was associated with increased levels of circulating nuclear autoantigens (P < 0.01), anti-double-stranded DNA autoantibodies (P < 0.01), tissue immune complex deposition (P < 0.05), expansion of myeloid cell subsets (P < 0.05), and enhanced B cell activation and plasma cell differentiation (P = 0.001). Moreover, in the absence of GSDMD, enhanced autoantigen generation was associated with increased local induction of cell death in vivo. CONCLUSION GSDMD negatively regulates autoantigen generation and immune dysregulation in response to tissue injury and may play previously unappreciated protective roles in systemic autoimmunity.
Collapse
Affiliation(s)
- Xinghao Wang
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, United States
| | - Luz P Blanco
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, United States
| | - Carmelo Carmona-Rivera
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, United States
| | - Shuichiro Nakabo
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, United States
| | - Hege L Pedersen
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, United States
| | - Zu-Xi Yu
- National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, United States
| | - Mariana J Kaplan
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, United States
| |
Collapse
|
376
|
Santocki M, Kolaczkowska E. On Neutrophil Extracellular Trap (NET) Removal: What We Know Thus Far and Why So Little. Cells 2020; 9:cells9092079. [PMID: 32932841 PMCID: PMC7565917 DOI: 10.3390/cells9092079] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/05/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022] Open
Abstract
Although neutrophil extracellular traps (NETs) were discovered only 16 years ago, they have already taken us from heaven to hell as we learned that apart from beneficial trapping of pathogens, they cause, or contribute to, numerous disorders. The latter is connected to their persistent presence in the blood or tissue, and we hardly know how they are removed in mild pathophysiological conditions and why their removal is impaired in multiple severe pathological conditions. Herein, we bring together all data available up till now on how NETs are cleared—from engaged cells, their phenotypes, to involved enzymes and molecules. Moreover, we hypothesize on why NET removal is challenged in multiple disorders and propose further directions for studies on NET removal as well as possible therapeutic strategies to have them cleared.
Collapse
|
377
|
Lu YJ, Wang YH, Sahu RS, Chen JP, Dash BS, Chung PJ, Yang HW, Chuang EY, Hwang TL. Mechanism of Nanoformulated Graphene Oxide-Mediated Human Neutrophil Activation. ACS APPLIED MATERIALS & INTERFACES 2020; 12:40141-40152. [PMID: 32845120 DOI: 10.1021/acsami.0c12490] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Understanding the molecular mechanisms of graphene oxide (GO)-based biomaterials is important for logical biomedical applications. Previous studies have revealed biointeractions between GO and immune effector cells, but the effects on neutrophils, crucial cells in the immune system, have not been thoroughly discussed. In this study, GO nanoformulations were synthesized with different functional groups, including GO, GO-carboxylated (GO-COOH), and PEGylated GO (GO-PEG), with different surface features, which were elucidated using imaging methods and surface-sensitive quantitative spectroscopic techniques, including atomic force microscopy (AFM), transmission electron microscopy (TEM), and X-ray photoemission spectroscopy (XPS). The GO-based nanoformulations elicited reactive oxygen species (ROS) generation and neutrophil extracellular trap (NET) formation in human neutrophils. Nanoformulated GO stimulates NET development via the formation of ROS. An endocytosis study revealed that nanoformulated GO facilitated internalization by neutrophils via macropinocytosis and actin-dependent phagocytosis. Importantly, calcium mobilization and phosphorylation proteins such as mitogen-activated protein kinases (extracellular signal-regulated kinase, c-Jun N-terminal kinase, and p38) and AKT were involved in the activation of neutrophils. These findings offer the first verification that nanoformulated GO exhibits direct effects on human neutrophils.
Collapse
Affiliation(s)
- Yu-Jen Lu
- Department of Neurosurgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Yi-Hsuan Wang
- Graduate Institute of Natural Products, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Rama Shanker Sahu
- Department of Agricultural Chemistry, National Taiwan University, Taipei 106, Taiwan
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Banendu Sunder Dash
- Department of Chemical and Materials Engineering, Chang Gung University, Taoyuan 33302, Taiwan
| | - Pei-Jen Chung
- Graduate Institute of Natural Products, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Hung-Wei Yang
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, International Ph.D. Program of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
- Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei 116, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 33302, Taiwan
- Department of Chemical Engineering, Ming-Chi University of Technology, New Taipei City 243, Taiwan
| |
Collapse
|
378
|
Soh KY, Loh JMS, Hall C, Proft T. Functional Analysis of Two Novel Streptococcus iniae Virulence Factors Using a Zebrafish Infection Model. Microorganisms 2020; 8:E1361. [PMID: 32899555 PMCID: PMC7564053 DOI: 10.3390/microorganisms8091361] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/30/2022] Open
Abstract
Streptococcus iniae is a major fish pathogen that contributes to large annual losses in the aquaculture industry, exceeding US$100 million. It is also reported to cause opportunistic infections in humans. We have recently identified two novel S. iniae virulence factors, an extracellular nuclease (SpnAi) and a secreted nucleotidase (S5nAi), and verified their predicted enzymatic activities using recombinant proteins. Here, we report the generation of green fluorescent S. iniae spnAi and s5nAi deletion mutants and their evaluation in a transgenic zebrafish infection model. Our results show nuclease and nucleotidase activities in S. iniae could be attributed to SpnAi and S5nAi, respectively. Consistent with this, larvae infected with the deletion mutants demonstrated enhanced survival and bacterial clearance, compared to those infected with wild-type (WT) S. iniae. Deletion of spnAi and s5nAi resulted in sustained recruitment of neutrophils and macrophages, respectively, to the site of infection. We also show that recombinant SpnAi is able to degrade neutrophil extracellular traps (NETs) isolated from zebrafish kidney tissue. Our results suggest that both enzymes play an important role in S. iniae immune evasion and might present potential targets for the development of therapeutic agents or vaccines.
Collapse
Affiliation(s)
- Kar Yan Soh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland 1142, New Zealand; (K.Y.S.); (J.M.S.L.)
- Maurice Wilkins Centre for Biomolecular Discoveries, The University of Auckland, Auckland 1142, New Zealand
| | - Jacelyn Mei San Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland 1142, New Zealand; (K.Y.S.); (J.M.S.L.)
- Maurice Wilkins Centre for Biomolecular Discoveries, The University of Auckland, Auckland 1142, New Zealand
| | - Christopher Hall
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland 1142, New Zealand; (K.Y.S.); (J.M.S.L.)
| | - Thomas Proft
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland 1142, New Zealand; (K.Y.S.); (J.M.S.L.)
- Maurice Wilkins Centre for Biomolecular Discoveries, The University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
379
|
Sreejit G, Nagareddy PR. Response by Sreejit and Nagareddy to Letter Regarding Article, "Neutrophil-Derived S100A8/A9 Amplify Granulopoiesis After Myocardial Infarction". Circulation 2020; 142:e125-e126. [PMID: 32866066 DOI: 10.1161/circulationaha.120.049408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Gopalkrishna Sreejit
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus
| | - Prabhakara R Nagareddy
- Division of Cardiac Surgery, Department of Surgery, Ohio State University Wexner Medical Center, Columbus
| |
Collapse
|
380
|
Reithofer M, Karacs J, Strobl J, Kitzmüller C, Polak D, Seif K, Kamalov M, Becker CFW, Greiner G, Schmetterer K, Stary G, Bohle B, Jahn-Schmid B. Alum triggers infiltration of human neutrophils ex vivo and causes lysosomal destabilization and mitochondrial membrane potential-dependent NET-formation. FASEB J 2020; 34:14024-14041. [PMID: 32860638 PMCID: PMC7589265 DOI: 10.1096/fj.202001413r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 01/30/2023]
Abstract
Aluminium salts have been used in vaccines for decades. However, the mechanisms underlying their adjuvant effect are still unclear. Neutrophils, the first immune cells at the injection site, can release cellular DNA together with granular material, so‐called neutrophil extracellular traps (NETs). In mice, NETs apparently play a role in aluminium hydroxide (alum)‐adjuvant immune response to vaccines. Although no experimental data exist, this effect is assumed to be operative also in humans. As a first step to verify this knowledge in humans, we demonstrate that the injection of alum particles into human skin biopsies ex vivo leads to similar tissue infiltration of neutrophils and NET‐formation. Moreover, we characterized the mechanism leading to alum‐induced NET‐release in human neutrophils as rapid, NADPH oxidase‐independent process involving charge, phagocytosis, phagolysosomal rupture, Ca2+‐flux, hyperpolarization of the mitochondrial membrane, and mitochondrial ROS. Extracellular flow and inhibition experiments suggested that no additional energy from oxidative phosphorylation or glycolysis is required for NET‐release. This study suggests a so far unappreciated role for neutrophils in the initial phase of immune responses to alum‐containing vaccines in humans and provides novel insights into bioenergetic requirements of NET‐formation.
Collapse
Affiliation(s)
- Manuel Reithofer
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Jasmine Karacs
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Johanna Strobl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Claudia Kitzmüller
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Dominika Polak
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Katharina Seif
- Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Meder Kamalov
- Institute of Biological Chemistry, Department of Chemistry, University of Vienna, Vienna, Austria
| | - Christian F W Becker
- Institute of Biological Chemistry, Department of Chemistry, University of Vienna, Vienna, Austria
| | - Georg Greiner
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Klaus Schmetterer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Georg Stary
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,CeMM Research Center for Molecular Medicine, Vienna, Austria
| | - Barbara Bohle
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Beatrice Jahn-Schmid
- Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
381
|
Li T, Peng R, Wang F, Hua L, Liu S, Han Z, Pei J, Pei S, Zhao Z, Jiang X, Chen X. Lysophosphatidic acid promotes thrombus stability by inducing rapid formation of neutrophil extracellular traps: A new mechanism of thrombosis. J Thromb Haemost 2020; 18:1952-1964. [PMID: 32291893 DOI: 10.1111/jth.14839] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 03/29/2020] [Accepted: 04/08/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lysophosphatidic acid (LPA), a bioactive phospholipid released by activated platelets, can induce platelet shape changes and aggregation, which may play an important role in thrombosis. In contrast, the interaction of LPA with neutrophils in thrombosis has not been studied. Recently, neutrophil extracellular traps (NETs) have been shown to bind plasma proteins and activate platelets, which promotes thrombosis. OBJECTIVES To investigate whether LPA could activate neutrophils to release NETs, predisposing to thrombosis and promoting thrombus stability. METHODS Levels of neutrophils, NETs, and LPA were detected in 56 participants. Immunofluorescence of NETs and autotaxin, the LPA-producing ectoenzyme, were performed. Induction of NETs and signaling pathways were explored in vitro. RESULTS Patients with acute pulmonary embolism showed elevated levels of neutrophils, NETs (dsDNA, MPO-DNA, citrullinated histone H3, and nucleosomes), LPA18:1, and LPA20:4. NETs were present in human intrapulmonary thrombi and were surrounded by autotaxin. LPA18:1 induced rapid release of NETs from human neutrophils via a peptidylarginine deiminase 4-dependent pathway. LPA-induced NETs provided a scaffolding for plasma protein binding and generated a tissue plasminogen activator (tPA)-resistant blood clot. Addition of deoxyribonuclease I to tPA significantly accelerated the lysis of clots and human intrapulmonary thrombi. Furthermore, LPA-induced NETs could activate platelets to release LPA. CONCLUSION This is the first study to implicate LPA in regulating the stability of thrombi by inducing rapid release of NETs in vitro and ex vivo, which could be a new mechanism of thrombosis. These findings provide new insight into the prevention and therapy of venous thromboembolic disease by targeting the LPA-NET signaling pathway.
Collapse
Affiliation(s)
- Tiewei Li
- Diagnostic Laboratory Service, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Rui Peng
- Diagnostic Laboratory Service, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Fang Wang
- Diagnostic Laboratory Service, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lu Hua
- Key Laboratory of Pulmonary Vascular Medicine & Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Sheng Liu
- Department of Cardiac Surgery, Adult Cardiac Surgery Center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhiyan Han
- Department of Anesthesiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jianqiu Pei
- Diagnostic Laboratory Service, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shengqiang Pei
- Diagnostic Laboratory Service, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhenwen Zhao
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Xin Jiang
- Department of Cardiology, Key Laboratory of Pulmonary Vascular Medicine, Peking Union Medical College Hospital, Chinese Academy Medical Sciences, Beijing, China
| | - Xi Chen
- Diagnostic Laboratory Service, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
382
|
Manda-Handzlik A, Bystrzycka W, Cieloch A, Glodkowska-Mrowka E, Jankowska-Steifer E, Heropolitanska-Pliszka E, Skrobot A, Muchowicz A, Ciepiela O, Wachowska M, Demkow U. Nitric oxide and peroxynitrite trigger and enhance release of neutrophil extracellular traps. Cell Mol Life Sci 2020; 77:3059-3075. [PMID: 31650185 PMCID: PMC7366602 DOI: 10.1007/s00018-019-03331-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/07/2019] [Accepted: 10/09/2019] [Indexed: 12/18/2022]
Abstract
Despite great interest, the mechanism of neutrophil extracellular traps (NETs) release is not fully understood and some aspects of this process, e.g. the role of reactive nitrogen species (RNS), still remain unclear. Therefore, our aim was to investigate the mechanisms underlying RNS-induced formation of NETs and contribution of RNS to NETs release triggered by various physiological and synthetic stimuli. The involvement of RNS in NETs formation was studied in primary human neutrophils and differentiated human promyelocytic leukemia cells (HL-60 cells). RNS (peroxynitrite and nitric oxide) efficiently induced NETs release and potentiated NETs-inducing properties of platelet activating factor and lipopolysaccharide. RNS-induced NETs formation was independent of autophagy and histone citrullination, but dependent on the activity of phosphoinositide 3-kinases (PI3K) and myeloperoxidase, as well as selective degradation of histones H2A and H2B by neutrophil elastase. Additionally, NADPH oxidase activity was required to release NETs upon stimulation with NO, as shown in NADPH-deficient neutrophils isolated from patients with chronic granulomatous disease. The role of RNS was further supported by increased RNS synthesis upon stimulation of NETs release with phorbol 12-myristate 13-acetate and calcium ionophore A23187. Scavenging or inhibition of RNS formation diminished NETs release triggered by these stimuli while scavenging of peroxynitrite inhibited NO-induced NETs formation. Our data suggest that RNS may act as mediators and inducers of NETs release. These processes are PI3K-dependent and ROS-dependent. Since inflammatory reactions are often accompanied by nitrosative stress and NETs formation, our studies shed a new light on possible mechanisms engaged in various immune-mediated conditions.
Collapse
Affiliation(s)
- Aneta Manda-Handzlik
- Department of Laboratory Medicine and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091, Warsaw, Poland
| | - Weronika Bystrzycka
- Department of Laboratory Medicine and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Zwirki i Wigury 61 Street, 02-091, Warsaw, Poland
| | - Adrianna Cieloch
- Department of Laboratory Medicine and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| | - Eliza Glodkowska-Mrowka
- Department of Laboratory Medicine and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
- The Finsen Laboratory, Faculty of Health Sciences, Rigshospitalet, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen, Denmark
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen, Denmark
- Faculty of Health Sciences, Danish Stem Cell Centre (DanStem), University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen, Denmark
- Department of Biology, The Bioinformatics Centre, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen, Denmark
| | - Ewa Jankowska-Steifer
- Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5 Street, 02-004, Warsaw, Poland
| | - Edyta Heropolitanska-Pliszka
- Department of Immunology, The Children's Memorial Health Institute, Aleja Dzieci Polskich 20, 04-730, Warsaw, Poland
| | - Agnieszka Skrobot
- Department of Laboratory Medicine and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| | - Angelika Muchowicz
- Department of Immunology, Medical University of Warsaw, Jana Nielubowicza 5 Street, 02-097, Warsaw, Poland
| | - Olga Ciepiela
- Department of Laboratory Diagnostics, Medical University of Warsaw, Banacha 1a Street, 02-097, Warsaw, Poland
| | - Malgorzata Wachowska
- Department of Laboratory Medicine and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland.
| | - Urszula Demkow
- Department of Laboratory Medicine and Clinical Immunology of Developmental Age, Medical University of Warsaw, Zwirki i Wigury 63a Street, 02-091, Warsaw, Poland
| |
Collapse
|
383
|
Neutrophil Extracellular Traps: Signaling Properties and Disease Relevance. Mediators Inflamm 2020; 2020:9254087. [PMID: 32774152 PMCID: PMC7407020 DOI: 10.1155/2020/9254087] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are characterized as extracellular DNA fibers comprised of histone and cytoplasmic granule proteins. NETs were first described as a form of innate response against pathogen invasion, which can capture pathogens, degrade bacterial toxic factors, and kill bacteria. Additionally, NETs also provide a scaffold for protein and cell binding. Protein binding to NETs further activate the coagulation system which participates in thrombosis. In addition, NETs also can damage the tissues due to the proteins they carry. Many studies have suggested that the excessive formation of NETs may contribute to a range of diseases, including thrombosis, atherosclerosis, autoimmune diseases, and sepsis. In this review, we describe the structure and components of NETs, models of NET formation, and detection methods. We also discuss the molecular mechanism of NET formation and their disease relevance. Modulation of NET formation may provide a new route for the prevention and treatment of releated human diseases.
Collapse
|
384
|
Locke M, Francis RJ, Tsaousi E, Longstaff C. Fibrinogen protects neutrophils from the cytotoxic effects of histones and delays neutrophil extracellular trap formation induced by ionomycin. Sci Rep 2020; 10:11694. [PMID: 32678135 PMCID: PMC7366688 DOI: 10.1038/s41598-020-68584-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/30/2020] [Indexed: 12/27/2022] Open
Abstract
Neutrophils are pivotal players in immune defence which includes a process of release of histones and DNA as neutrophil extracellular traps (NETs). Histones, while toxic to invading pathogens, also kill host cells, including neutrophils. Bacteria have evolved mechanisms to escape neutrophils, including the secretion of leucocidins (e.g. ionomycin). Live cell video microscopy showed how fibrinogen and fibrin influence NETosis and neutrophil responses to extracellular histones. Histones were rapidly lethal to neutrophils after binding to cells, but formation of fibrinogen/fibrin-histone aggregates prevented cell death. Histone cytotoxicity was also reduced by citrullination by peptidyl arginine deiminase 4, or digestion by serine proteases. Ionomycin and phorbol 12-myristate 13 acetate (PMA) are used to trigger NETosis. Fibrinogen was responsible for a second distinct mechanism of neutrophil protection after treatment with ionomycin. Fibrinogen clustered on the surface of ionomycin-stimulated neutrophils to delay NETosis; and blocking the β integrin receptor, αMβ2, abolished fibrinogen protection. Fibrinogen did not bind to or protect neutrophils stimulated with PMA. Fibrinogen is an acute phase protein that will protect exposed cells from damaging circulating histones or leucocidins; but fibrinogen depletion/consumption, as in trauma or sepsis will reduce protection. It is necessary to consider the role of fibrinogen in NETosis.
Collapse
Affiliation(s)
- Matthew Locke
- Biotherapeutics, National Institute for Biological Standards and Control, S Mimms, Herts, UK
| | - Robert J Francis
- Biological Imaging Group, Analytical Biological Sciences, National Institute for Biological Standards and Control, S Mimms, Herts, UK
| | - Evgenia Tsaousi
- Biotherapeutics, National Institute for Biological Standards and Control, S Mimms, Herts, UK.,School of Biological Sciences, University of Essex, Colchester, UK
| | - Colin Longstaff
- Biotherapeutics, National Institute for Biological Standards and Control, S Mimms, Herts, UK.
| |
Collapse
|
385
|
Filep JG, Ariel A. Neutrophil heterogeneity and fate in inflamed tissues: implications for the resolution of inflammation. Am J Physiol Cell Physiol 2020; 319:C510-C532. [PMID: 32667864 DOI: 10.1152/ajpcell.00181.2020] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neutrophils are polymorphonuclear leukocytes that play a central role in host defense against infection and tissue injury. They are rapidly recruited to the inflamed site and execute a variety of functions to clear invading pathogens and damaged cells. However, many of their defense mechanisms are capable of inflicting collateral tissue damage. Neutrophil-driven inflammation is a unifying mechanism underlying many common diseases. Efficient removal of neutrophils from inflammatory loci is critical for timely resolution of inflammation and return to homeostasis. Accumulating evidence challenges the classical view that neutrophils represent a homogeneous population and that halting neutrophil influx is sufficient to explain their rapid decline within inflamed loci during the resolution of protective inflammation. Hence, understanding the mechanisms that govern neutrophil functions and their removal from the inflammatory locus is critical for minimizing damage to the surrounding tissue and for return to homeostasis. In this review, we briefly address recent advances in characterizing neutrophil phenotypic and functional heterogeneity and the molecular mechanisms that determine the fate of neutrophils within inflammatory loci and the outcome of the inflammatory response. We also discuss how these mechanisms may be harnessed as potential therapeutic targets to facilitate resolution of inflammation.
Collapse
Affiliation(s)
- János G Filep
- Department of Pathology and Cell Biology, University of Montreal and Research Center, Maisonneuve-Rosemont Hospital, Montreal, Quebec, Canada
| | - Amiram Ariel
- Departmentof Biology and Human Biology, University of Haifa, Haifa, Israel
| |
Collapse
|
386
|
Abstract
Neutrophils are critical to innate immunity, including host defense against bacterial and fungal infections. They achieve their host defense role by phagocytosing pathogens, secreting their granules full of cytotoxic enzymes, or expelling neutrophil extracellular traps (NETs) during the process of NETosis. NETs are weblike DNA structures decorated with histones and antimicrobial proteins released by activated neutrophils. Initially described as a means for neutrophils to neutralize pathogens, NET release also occurs in sterile inflammation, promotes thrombosis, and can mediate tissue damage. To effectively manipulate this double-edged sword to fight a particular disease, researchers must work toward understanding the mechanisms driving NETosis. Such understanding would allow the generation of new drugs to promote or prevent NETosis as needed. While knowledge regarding the (patho)physiological roles of NETosis is accumulating, little is known about the cellular and biophysical bases of this process. In this review, we describe and discuss our current knowledge of the molecular, cellular, and biophysical mechanisms mediating NET release as well as open questions in the field.
Collapse
Affiliation(s)
- Hawa Racine Thiam
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland 20892, USA; ,
| | - Siu Ling Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232
| | - Denisa D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Clare M Waterman
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland 20892, USA; ,
| |
Collapse
|
387
|
Burgener SS, Schroder K. Neutrophil Extracellular Traps in Host Defense. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037028. [PMID: 31767647 DOI: 10.1101/cshperspect.a037028] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neutrophils are produced in the bone marrow and then patrol blood vessels from which they can be rapidly recruited to a site of infection. Neutrophils bind, engulf, and efficiently kill invading microbes via a suite of defense mechanisms. Diverse extracellular and intracellular microbes induce neutrophils to extrude neutrophil extracellular traps (NETs) through the process of NETosis. Here, we review the signaling mechanisms and cell biology underpinning the key NETosis pathways during infection and the antimicrobial functions of NETs in host defense.
Collapse
Affiliation(s)
- Sabrina Sofia Burgener
- Institute for Molecular Bioscience (IMB), and IMB Centre for Inflammation and Disease Research, The University of Queensland, St Lucia 4072, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), and IMB Centre for Inflammation and Disease Research, The University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
388
|
Xu D, Lin Y, Shen J, Zhang J, Wang J, Zhang Y, Zhang H, Ning L, Liu P, Li S, Zeng H, Lin J, Yu C. Overproduced bone marrow neutrophils in collagen-induced arthritis are primed for NETosis: An ignored pathological cell involving inflammatory arthritis. Cell Prolif 2020; 53:e12824. [PMID: 32567730 PMCID: PMC7377937 DOI: 10.1111/cpr.12824] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/31/2020] [Accepted: 04/19/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Bone marrow edema is a universal manifestation of rheumatoid arthritis (RA), and its pathological essence is a bone marrow lesion (BML) formed by various bone marrow (BM) immune cells. Neutrophils play an important role in inflammatory arthritis, but the role and mechanism of neutrophils in BML are not clear. MATERIALS AND METHODS Granulocyte colony-stimulating factor (G-CSF) -/- mice and wild type (WT) C57BL/6 mice were immunized for collagen-induced arthritis (CIA). Histological scores of arthritis were evaluated. Immunohistochemistry staining with anti-Ly6G was conducted. Neutrophil extracellular traps (NETs) in joint sections were determined by immunofluorescence staining. BM neutrophils were isolated for flow cytometry and NETosis induction in vitro. RESULTS Histological study showed significant neutrophil infiltrations in BML of CIA mice. Inhibition of BM neutrophil production by G-CSF knock out can obstruct the induction of BML and CIA. In addition to abundant infiltrated NETs intra-articular, remarkable NETosis primed BM neutrophils were infiltrated in BML of CIA mice, which was positively related to bone erosion. Neutrophils derived from G-CSF-/- mice have diminished ability of NETs formation in vitro, while G-CSF induction can enhance its capacity of NETs formation. CONCLUSIONS We propose for the first time that the overproduced BM neutrophils in CIA mice are primed for NETosis in a G-CSF dependent manner, and these pathogenic cells may have an important role in inflammatory arthritis. Blocking this pathological process could be a potential strategy for the treatment of RA.
Collapse
Affiliation(s)
- Danyi Xu
- Department of RheumatologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Yiming Lin
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Jinming Shen
- Department of OrthopaedicsThe First Affiliated HospitalZhejiang University of Traditional Chinese MedicineHangzhouChina
| | - Jie Zhang
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Jinghua Wang
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Yuwei Zhang
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Hong Zhang
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Longgui Ning
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Peihao Liu
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Sha Li
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Hang Zeng
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Jin Lin
- Department of RheumatologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Chaohui Yu
- Department of GastroenterologyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
389
|
Demarco B, Chen KW, Broz P. Cross talk between intracellular pathogens and cell death. Immunol Rev 2020; 297:174-193. [PMID: 32567717 DOI: 10.1111/imr.12892] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023]
Abstract
Infections with bacterial pathogens often results in the initiation of programmed cell death as part of the host innate immune defense, or as a bacterial virulence strategy. Induction of host cell death is controlled by an elaborate network of innate immune and cell death signaling pathways and manifests in different morphologically and functionally distinct forms of death, such as apoptosis, necroptosis, NETosis and pyroptosis. The mechanism by which host cell death restricts bacterial replication is highly cell-type and context depended, but its physiological importance is highlighted the diversity of strategies bacterial pathogens use to avoid induction of cell death or to block cell death signaling pathways. In this review, we discuss the latest insights into how bacterial pathogens elicit and manipulate cell death signaling, how different forms of cell death kill or restrict bacteria and how cell death and innate immune pathway cross talk to guard against pathogen-induced inhibition of host cell death.
Collapse
Affiliation(s)
- Benjamin Demarco
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Kaiwen W Chen
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Petr Broz
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
390
|
Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M, Madison JA, Blair C, Weber A, Barnes BJ, Egeblad M, Woods RJ, Kanthi Y, Knight JS. Neutrophil extracellular traps in COVID-19. JCI Insight 2020; 5:138999. [PMID: 32329756 PMCID: PMC7308057 DOI: 10.1172/jci.insight.138999] [Citation(s) in RCA: 778] [Impact Index Per Article: 155.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/23/2020] [Indexed: 02/06/2023] Open
Abstract
In severe cases of coronavirus disease 2019 (COVID-19), viral pneumonia progresses to respiratory failure. Neutrophil extracellular traps (NETs) are extracellular webs of chromatin, microbicidal proteins, and oxidant enzymes that are released by neutrophils to contain infections. However, when not properly regulated, NETs have the potential to propagate inflammation and microvascular thrombosis - including in the lungs of patients with acute respiratory distress syndrome. We now report that sera from patients with COVID-19 have elevated levels of cell-free DNA, myeloperoxidase-DNA (MPO-DNA), and citrullinated histone H3 (Cit-H3); the latter 2 are specific markers of NETs. Highlighting the potential clinical relevance of these findings, cell-free DNA strongly correlated with acute-phase reactants, including C-reactive protein, D-dimer, and lactate dehydrogenase, as well as absolute neutrophil count. MPO-DNA associated with both cell-free DNA and absolute neutrophil count, while Cit-H3 correlated with platelet levels. Importantly, both cell-free DNA and MPO-DNA were higher in hospitalized patients receiving mechanical ventilation as compared with hospitalized patients breathing room air. Finally, sera from individuals with COVID-19 triggered NET release from control neutrophils in vitro. Future studies should investigate the predictive power of circulating NETs in longitudinal cohorts and determine the extent to which NETs may be novel therapeutic targets in severe COVID-19.
Collapse
Affiliation(s)
- Yu Zuo
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Srilakshmi Yalavarthi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hui Shi
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Division of Rheumatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kelsey Gockman
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Melanie Zuo
- Division of Geriatric and Palliative Medicine and
| | - Jacqueline A. Madison
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Christopher Blair
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Andrew Weber
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Northwell Health, New York, New York, USA
| | - Betsy J. Barnes
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Departments of Molecular Medicine and Pediatrics, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Robert J. Woods
- Division of Infectious Disease, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yogendra Kanthi
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Division of Cardiology, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
391
|
Zhao X, Yang L, Chang N, Hou L, Zhou X, Yang L, Li L. Neutrophils undergo switch of apoptosis to NETosis during murine fatty liver injury via S1P receptor 2 signaling. Cell Death Dis 2020; 11:379. [PMID: 32424179 PMCID: PMC7235026 DOI: 10.1038/s41419-020-2582-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 01/18/2023]
Abstract
Inappropriate neutrophil infiltration and subsequent neutrophil extracellular trap (NET) formation have been confirmed to be involved in chronic inflammatory conditions. Fatty liver disease is an increasingly severe health problem worldwide and currently considered the most common cause of chronic liver disease. Sphingosine 1-phosphate (S1P), a product of membrane sphingolipid metabolism, regulates vital physiological and pathological actions by inducing infiltration and activation of various cell types through S1P receptors (S1PRs). Here, we seek to determine the S1PR-mediated effects on neutrophil activation during chronic liver inflammation. In this study, NETs are detected in the early stage of methionine-choline-deficient and a high-fat (MCDHF) diet-induced liver injury. NET depletion by deoxyribonuclease I intraperitoneal injection significantly protects liver from MCDHF-induced liver injury in vivo. Meanwhile, we show that levels of myeloperoxidase-DNA complex (NET marker) in the serum present positive correlation with sphingosine kinase1 (S1P rate-limiting enzyme) messenger RNA expression or S1P levels in the injured liver of MCDHF-fed mice. In vitro, S1PR2 participates in the redirection of neutrophil apoptosis to NETosis via Gαi/o, extracellular signal-regulated kinase, p38 mitogen-activated protein kinase, and reactive oxygen species signaling pathways. Moreover, S1PR2 knockdown in MCDHF-fed mice by S1PR2-siRNA intravenous injection significantly inhibits NET formation in damaged liver tissue and then alleviates hepatic inflammation and fibrosis. Conclusion: In the early stage of fatty liver disease, S1PR2-mediated neutrophil activation plays an important role in the evolvement of liver injury.
Collapse
Affiliation(s)
- Xinhao Zhao
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, 100069, Beijing, China
| | - Le Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, 100069, Beijing, China
| | - Na Chang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, 100069, Beijing, China
| | - Lei Hou
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, 100069, Beijing, China
| | - Xuan Zhou
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, 100069, Beijing, China
| | - Lin Yang
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, 100069, Beijing, China
| | - Liying Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, 100069, Beijing, China.
| |
Collapse
|
392
|
Sollberger G, Streeck R, Apel F, Caffrey BE, Skoultchi AI, Zychlinsky A. Linker histone H1.2 and H1.4 affect the neutrophil lineage determination. eLife 2020; 9:52563. [PMID: 32391789 PMCID: PMC7250579 DOI: 10.7554/elife.52563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 05/08/2020] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are important innate immune cells that tackle invading pathogens with different effector mechanisms. They acquire this antimicrobial potential during their maturation in the bone marrow, where they differentiate from hematopoietic stem cells in a process called granulopoiesis. Mature neutrophils are terminally differentiated and short-lived with a high turnover rate. Here, we show a critical role for linker histone H1 on the differentiation and function of neutrophils using a genome-wide CRISPR/Cas9 screen in the human cell line PLB-985. We systematically disrupted expression of somatic H1 subtypes to show that individual H1 subtypes affect PLB-985 maturation in opposite ways. Loss of H1.2 and H1.4 induced an eosinophil-like transcriptional program, thereby negatively regulating the differentiation into the neutrophil lineage. Importantly, H1 subtypes also affect neutrophil differentiation and the eosinophil-directed bias of murine bone marrow stem cells, demonstrating an unexpected subtype-specific role for H1 in granulopoiesis.
Collapse
Affiliation(s)
- Gabriel Sollberger
- Max Planck Institute for Infection Biology, Department of Cellular Microbiology, Berlin, Germany.,University of Dundee, School of Life Sciences, Division of Cell Signalling and Immunology, Dundee, United Kingdom
| | - Robert Streeck
- Max Planck Institute for Infection Biology, Department of Cellular Microbiology, Berlin, Germany.,Institut für Biologie, Humboldt Universität zu Berlin, Berlin, Germany
| | - Falko Apel
- Max Planck Institute for Infection Biology, Department of Cellular Microbiology, Berlin, Germany.,Institut für Biologie, Humboldt Universität zu Berlin, Berlin, Germany
| | | | - Arthur I Skoultchi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, United States
| | - Arturo Zychlinsky
- Max Planck Institute for Infection Biology, Department of Cellular Microbiology, Berlin, Germany
| |
Collapse
|
393
|
Yin K, Cui Y, Qu Y, Zhang J, Zhang H, Lin H. Hydrogen sulfide upregulates miR-16-5p targeting PiK3R1 and RAF1 to inhibit neutrophil extracellular trap formation in chickens. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 194:110412. [PMID: 32155482 DOI: 10.1016/j.ecoenv.2020.110412] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 06/10/2023]
Abstract
Hydrogen sulfide (H2S) is a toxic air pollutant that causes immune damage. Recent studies have found that neutrophil extracellular trap (NET) formation is one way in which neutrophils exert immune functions. In addition, the formation of NETs is also related to thrombosis and autoimmune diseases. Recent studies have shown that miRNAs are involved in the regulation of a variety of pathophysiological processes. Here, we investigated the role of H2S in regulating the formation of NETs by affecting miR-16-5p. Our study established an in vitro H2S exposure model for neutrophils using phorbol-myristate-acetate (PMA) to induce NET formation. We observed the morphological changes of cells with scanning electron microscopy and fluorescence microscopy. Then, the content of extracellular DNA and the expression of MPO and NE in each group were detected. The results showed that H2S inhibited the formation of NETs. The expression of miR-16-5p and its target genes PiK3R1 and RAF1 was then measured by qRT-PCR. H2S upregulated miR-16-5p and inhibited expression of the target genes PiK3R1 and RAF1, and it subsequently inhibited the Pi3K/AKT and ERK pathways and decreased respiratory burst levels. Furthermore, H2S attenuated inositol 1,4,5-trisphosphate receptor (IP3R)-mediated endoplasmic reticulum calcium outflow as well as autophagy caused by PMA. This study enriches H2S immunotoxicity research and provides a possible solution for the treatment of NET-related diseases.
Collapse
Affiliation(s)
- Kai Yin
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Number 2, Yuanmingyuan West Road, Haidian District, Beijing, 100193, PR China; College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yuan Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Yingying Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Jinxi Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Hongfu Zhang
- Institute of Animal Sciences of Chinese Academy of Agricultural Sciences, Number 2, Yuanmingyuan West Road, Haidian District, Beijing, 100193, PR China; State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100193, PR China.
| | - Hongjin Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| |
Collapse
|
394
|
Tsourouktsoglou TD, Warnatsch A, Ioannou M, Hoving D, Wang Q, Papayannopoulos V. Histones, DNA, and Citrullination Promote Neutrophil Extracellular Trap Inflammation by Regulating the Localization and Activation of TLR4. Cell Rep 2020; 31:107602. [DOI: 10.1016/j.celrep.2020.107602] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 12/13/2019] [Accepted: 04/10/2020] [Indexed: 12/29/2022] Open
|
395
|
Yousefi S, Simon D, Stojkov D, Karsonova A, Karaulov A, Simon HU. In vivo evidence for extracellular DNA trap formation. Cell Death Dis 2020; 11:300. [PMID: 32355207 PMCID: PMC7193637 DOI: 10.1038/s41419-020-2497-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/01/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Extracellular DNA trap formation is a cellular function of neutrophils, eosinophils, and basophils that facilitates the immobilization and killing of invading microorganisms in the extracellular milieu. To form extracellular traps, granulocytes release a scaffold consisting of mitochondrial DNA in association with granule proteins. As we understand more about the molecular mechanism for the formation of extracellular DNA traps, the in vivo function of this phenomenon under pathological conditions remains an enigma. In this article, we critically review the literature to summarize the evidence for extracellular DNA trap formation under in vivo conditions. Extracellular DNA traps have not only been detected in infectious diseases but also in chronic inflammatory diseases, as well as in cancer. While on the one hand, extracellular DNA traps clearly exhibit an important function in host defense, it appears that they can also contribute to the maintenance of inflammation and metastasis, suggesting that they may represent an interesting drug target for such pathological conditions.
Collapse
Affiliation(s)
- Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Antonina Karsonova
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia.
| |
Collapse
|
396
|
Meyer D, Telele S, Zelená A, Gillen AJ, Antonucci A, Neubert E, Nißler R, Mann FA, Erpenbeck L, Boghossian AA, Köster S, Kruss S. Transport and programmed release of nanoscale cargo from cells by using NETosis. NANOSCALE 2020; 12:9104-9115. [PMID: 32286598 DOI: 10.1039/d0nr00864h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Cells can take up nanoscale materials, which has important implications for understanding cellular functions, biocompatibility as well as biomedical applications. Controlled uptake, transport and triggered release of nanoscale cargo is one of the great challenges in biomedical applications of nanomaterials. Here, we study how human immune cells (neutrophilic granulocytes, neutrophils) take up nanomaterials and program them to release this cargo after a certain time period. For this purpose, we let neutrophils phagocytose DNA-functionalized single-walled carbon nanotubes (SWCNTs) in vitro that fluoresce in the near infrared (980 nm) and serve as sensors for small molecules. Cells still migrate, follow chemical gradients and respond to inflammatory signals after uptake of the cargo. To program release, we make use of neutrophil extracellular trap formation (NETosis), a novel cell death mechanism that leads to chromatin swelling, subsequent rupture of the cellular membrane and release of the cell's whole content. By using the process of NETosis, we can program the time point of cargo release via the initial concentration of stimuli such as phorbol 12-myristate-13-acetate (PMA) or lipopolysaccharide (LPS). At intermediate stimulation, cells continue to migrate, follow gradients and surface cues for around 30 minutes and up to several hundred micrometers until they stop and release the SWCNTs. The transported and released SWCNT sensors are still functional as shown by subsequent detection of the neurotransmitter dopamine and reactive oxygen species (H2O2). In summary, we hijack a biological process (NETosis) and demonstrate how neutrophils transport and release functional nanomaterials.
Collapse
Affiliation(s)
- Daniel Meyer
- Institute of Physical Chemistry, Göttingen University, 37077 Göttingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
397
|
Döring Y, Libby P, Soehnlein O. Neutrophil Extracellular Traps Participate in Cardiovascular Diseases: Recent Experimental and Clinical Insights. Circ Res 2020; 126:1228-1241. [PMID: 32324499 PMCID: PMC7185047 DOI: 10.1161/circresaha.120.315931] [Citation(s) in RCA: 240] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neutrophil extracellular traps (NETs) have recently emerged as a newly recognized contributor to venous and arterial thrombosis. These strands of DNA extruded by activated or dying neutrophils, decorated with various protein mediators, become solid-state reactors that can localize at the critical interface of blood with the intimal surface of diseased arteries and propagate and amplify the regional injury. NETs thus furnish a previously unsuspected link between inflammation, innate immunity, thrombosis, oxidative stress, and cardiovascular diseases. In response to disease-relevant stimuli, neutrophils undergo a specialized series of reactions that culminate in NET formation. DNA derived from either nuclei or mitochondria can contribute to NET formation. The DNA liberated from neutrophils forms a reticular mesh that resembles morphologically a net, rendering the acronym NETs particularly appropriate. The DNA backbone of NETs not only presents intrinsic neutrophil proteins (eg, MPO [myeloperoxidase] and various proteinases) but can gather other proteins found in blood (eg, tissue factor procoagulant). This review presents current concepts of neutrophil biology, the triggers to and mechanisms of NET formation, and the contribution of NETs to atherosclerosis and to thrombosis. We consider the use of markers of NETs in clinical studies. We aim here to integrate critically the experimental literature with the growing body of clinical information regarding NETs.
Collapse
Affiliation(s)
- Yvonne Döring
- Division of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, Bern, Switzerland
- Institute of Cardiovascular Prevention, Department of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Oliver Soehnlein
- Institute of Cardiovascular Prevention, Department of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Physiology and Pharmacology (FyFA), Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
398
|
Zuo Y, Yalavarthi S, Shi H, Gockman K, Zuo M, Madison JA, Blair C, Weber A, Barnes BJ, Egeblad M, Woods RJ, Kanthi Y, Knight JS. Neutrophil extracellular traps (NETs) as markers of disease severity in COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020. [PMID: 32511633 DOI: 10.1101/2020.04.09.20059626] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In severe cases of coronavirus disease 2019 (COVID-19), viral pneumonia progresses to respiratory failure. Neutrophil extracellular traps (NETs) are extracellular webs of chromatin, microbicidal proteins, and oxidant enzymes that are released by neutrophils to contain infections. However, when not properly regulated, NETs have potential to propagate inflammation and microvascular thrombosis, including in the lungs of patients with acute respiratory distress syndrome. While elevated levels of blood neutrophils predict worse outcomes in COVID-19, the role of NETs has not been investigated. We now report that sera from patients with COVID-19 (n=50 patients, n=84 samples) have elevated levels of cell-free DNA, myeloperoxidase(MPO)-DNA, and citrullinated histone H3 (Cit-H3); the latter two are highly specific markers of NETs. Highlighting the potential clinical relevance of these findings, cell-free DNA strongly correlated with acute phase reactants including C-reactive protein, D-dimer, and lactate dehydrogenase, as well as absolute neutrophil count. MPO-DNA associated with both cell-free DNA and absolute neutrophil count, while Cit-H3 correlated with platelet levels. Importantly, both cell-free DNA and MPO-DNA were higher in hospitalized patients receiving mechanical ventilation as compared with hospitalized patients breathing room air. Finally, sera from individuals with COVID-19 triggered NET release from control neutrophils in vitro. In summary, these data reveal high levels of NETs in many patients with COVID-19, where they may contribute to cytokine release and respiratory failure. Future studies should investigate the predictive power of circulating NETs in longitudinal cohorts, and determine the extent to which NETs may be novel therapeutic targets in severe COVID-19.
Collapse
|
399
|
Lehman HK, Segal BH. The role of neutrophils in host defense and disease. J Allergy Clin Immunol 2020; 145:1535-1544. [PMID: 32283205 DOI: 10.1016/j.jaci.2020.02.038] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/15/2020] [Accepted: 02/05/2020] [Indexed: 12/21/2022]
Abstract
Neutrophils, the most abundant circulating leukocyte, are critical for host defense. Granulopoiesis is under the control of transcriptional factors and culminates in mature neutrophils with a broad armamentarium of antimicrobial pathways. These pathways include nicotinamide adenine dinucleotide phosphate oxidase, which generates microbicidal reactive oxidants, and nonoxidant pathways that target microbes through several mechanisms. Activated neutrophils can cause or worsen tissue injury, underscoring the need for calibration of activation and resolution of inflammation when infection has been cleared. Acquired neutrophil disorders are typically caused by cytotoxic chemotherapy or immunosuppressive agents. Primary neutrophil disorders typically result from disabling mutations of individual genes that result in impaired neutrophil number or function, and provide insight into basic mechanisms of neutrophil biology. Neutrophils can also be activated by noninfectious causes, including trauma and cellular injury, and can have off-target effects in which pathways that typically defend against infection exacerbate injury and disease. These off-target effects include acute organ injury, autoimmunity, and variable effects on the tumor microenvironment that can limit or worsen tumor progression. A greater understanding of neutrophil plasticity in these conditions is likely to pave the way to new therapeutic approaches.
Collapse
Affiliation(s)
- Heather K Lehman
- Division of Allergy/Immunology & Rheumatology, Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Brahm H Segal
- Roswell Park Comprehensive Cancer Center, University at Buffalo, Buffalo, NY.
| |
Collapse
|
400
|
Gordon RA, Tilstra JS, Marinov A, Nickerson KM, Bastacky SI, Shlomchik MJ. Murine lupus is neutrophil elastase-independent in the MRL.Faslpr model. PLoS One 2020; 15:e0226396. [PMID: 32243431 PMCID: PMC7122749 DOI: 10.1371/journal.pone.0226396] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/16/2020] [Indexed: 12/18/2022] Open
Abstract
Loss of tolerance to nuclear antigens and multisystem tissue destruction is a hallmark of systemic lupus erythematosus (SLE). Although the source of autoantigen in lupus remains elusive, a compelling hypothetical source is dead cell debris that drives autoimmune activation. Prior reports suggest that neutrophil extracellular traps (NETs) and their associated death pathway, NETosis, are sources of autoantigen in SLE. However, others and we have shown that inhibition of NETs by targeting the NADPH oxidase complex and peptidylarginine deiminase 4 (PADI4) did not ameliorate disease in spontaneous murine models of SLE. Furthermore, myeloperoxidase and PADI4 deletion did not inhibit induced lupus. Since NET formation may occur independently of any one mediator, to address this controversy, we genetically deleted an additional important mediator of NETs and neutrophil effector function, neutrophil elastase (ELANE), in the MRL.Faslpr model of SLE. ELANE deficiency, and by extension ELANE-dependent NETs, had no effect on SLE nephritis, dermatitis, anti-self response, or immune composition in MRL.Faslpr mice. Taken together with prior data from our group and others, these data further challenge the paradigm that NETs and neutrophils are pathogenic in SLE.
Collapse
Affiliation(s)
- Rachael A. Gordon
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Jeremy S. Tilstra
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Anthony Marinov
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Kevin M. Nickerson
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Sheldon I. Bastacky
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Mark J. Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|