101
|
Takizawa S, Lee Y, Jacob A, Aziz M, Wang P. Neutrophil trogocytosis during their trans-endothelial migration: role of extracellular CIRP. Mol Med 2022; 28:91. [PMID: 35941574 PMCID: PMC9358840 DOI: 10.1186/s10020-022-00515-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022] Open
Abstract
Background Neutrophils are the most abundant innate immune cells in the circulating blood, and they act as the first responder against bacterial and fungal infection. However, accumulation of activated neutrophils can cause severe inflammation and tissue damage. Recently, neutrophil trogocytosis or membrane transfer with neighboring cells was reported to modulate immune responses. Extracellular cold-inducible RNA binding protein (eCIRP) is a newly identified damage-associated molecular pattern (DAMP). eCIRP can activate neutrophils to be more pro-inflammatory. This study aimed to identify the role of eCIRP in neutrophil trogocytosis during their trans-endothelial migration. Methods A trans-endothelial migration (TEM) assay using bone marrow neutrophils and mouse primary lung vascular endothelial cells was conducted using transwell chambers and neutrophil trogocytosis was assessed in vitro. In an in vivo mouse model of acute lung injury, neutrophil trogocytosis was assessed from bronchoalveolar lavage fluid. Results In TEM assay, the trogocytosis of neutrophils occurred during trans-endothelial migration and eCIRP significantly increased the percentage of these neutrophils. The trogocytosed neutrophils acquired the endothelial membrane containing junctional adhesion molecule-C (JAM-C) and VE-cadherin, and these membrane patches were polarized by Mac-1 binding. Furthermore, eCIRP-induced JAM-C positive trogocytosed neutrophils are more pro-inflammatory than the JAM-C negative counterpart. JAM-C positive trogocytosed neutrophils were also observed in the bronchoalveolar lavage fluid of a mouse model of acute lung injury. Conclusion These data suggest that during the paracellular trans-endothelial migration of neutrophils in response to inflammation, eCIRP induces trogocytosis of neutrophils, and the trogocytosed neutrophils exhibit an exaggerated pro-inflammatory phenotype promoting acute lung injury. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00515-3.
Collapse
Affiliation(s)
- Satoshi Takizawa
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Yongchan Lee
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
| | - Asha Jacob
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.,Departments of Molecular Medicine and Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.,Departments of Molecular Medicine and Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA. .,Departments of Molecular Medicine and Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA.
| |
Collapse
|
102
|
Braun B, Laib Sampaio K, Kuderna AK, Widmann M, Sinzger C. Viral and Cellular Factors Contributing to the Hematogenous Dissemination of Human Cytomegalovirus via Polymorphonuclear Leukocytes. Viruses 2022; 14:v14071561. [PMID: 35891541 PMCID: PMC9323586 DOI: 10.3390/v14071561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 02/06/2023] Open
Abstract
Polymorphonuclear leukocytes (PMNs) presumably transmit human cytomegalovirus (HCMV) between endothelial cells in blood vessels and thereby facilitate spread to peripheral organs. We aimed to identify viral components that contribute to PMN-mediated transmission and test the hypothesis that cellular adhesion molecules shield transmission sites from entry inhibitors. Stop codons were introduced into the genome of HCMV strain Merlin to delete pUL74 of the trimeric and pUL128 of the pentameric glycoprotein complex and the tegument proteins pp65 and pp71. Mutants were analyzed regarding virus uptake by PMNs and transfer of infection to endothelial cells. Cellular adhesion molecules were evaluated for their contribution to virus transmission using function-blocking antibodies, and hits were further analyzed regarding shielding against inhibitors of virus entry. The viral proteins pUL128, pp65, and pp71 were required for efficient PMN-mediated transmission, whereas pUL74 was dispensable. On the cellular side, the blocking of the αLβ2-integrin LFA-1 reduced virus transfer by 50% and allowed entry inhibitors to reduce it further by 30%. In conclusion, these data show that PMN-mediated transmission depends on the pentameric complex and an intact tegument and supports the idea of a virological synapse that promotes this dissemination mode both directly and via immune evasion.
Collapse
|
103
|
Structure modifications of 2-phenylquinoline by Aspergillus genera produce novel derivatives with potent leishmanicidal and anti-inflammatory properties. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
104
|
Integrin Regulators in Neutrophils. Cells 2022; 11:cells11132025. [PMID: 35805108 PMCID: PMC9266208 DOI: 10.3390/cells11132025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/17/2022] [Accepted: 06/22/2022] [Indexed: 02/01/2023] Open
Abstract
Neutrophils are the most abundant leukocytes in humans and are critical for innate immunity and inflammation. Integrins are critical for neutrophil functions, especially for their recruitment to sites of inflammation or infections. Integrin conformational changes during activation have been heavily investigated but are still not fully understood. Many regulators, such as talin, Rap1-interacting adaptor molecule (RIAM), Rap1, and kindlin, are critical for integrin activation and might be potential targets for integrin-regulating drugs in treating inflammatory diseases. In this review, we outline integrin activation regulators in neutrophils with a focus on the above critical regulators, as well as newly discovered modulators that are involved in integrin activation.
Collapse
|
105
|
Wu M, Liang Y, Zhang X. Changes in Pulmonary Microenvironment Aids Lung Metastasis of Breast Cancer. Front Oncol 2022; 12:860932. [PMID: 35719975 PMCID: PMC9204317 DOI: 10.3389/fonc.2022.860932] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Breast cancer has become the most common malignant disease in the world according to the International Agency for Research on Cancer (IARC), and the most critical cause of death is distant metastasis. The lung is the extremely common visceral site for breast cancer metastasis. Lung metastasis of breast cancer is not only dependent on the invasive ability of the tumor itself, but also closely relates to the pulmonary microenvironment. In the progression of breast cancer, the formation of specific microenvironment in lungs can provide suitable conditions for the metastasis of breast cancer. Pulmonary inflammatory response, angiogenesis, extracellular matrix remodeling, some chemotherapeutic agents and so on all play important roles in the formation of the pulmonary microenvironment. This review highlights recent findings regarding the alterations of pulmonary microenvironment in lung metastasis of breast cancer, with a focus on various cells and acellular components.
Collapse
Affiliation(s)
- Meimei Wu
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Jiangmen, China
| | - Yanfang Liang
- Department of Pathology, Dongguan Hospital Affiliated to Jinan University, Binhaiwan Central Hospital of Dongguan, Dongguan, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Key Laboratory of Clinical Biobanks and Translational Research, Jiangmen Central Hospital, Jiangmen, China.,Dongguan Key Laboratory of Medical Bioactive Molecular Developmental and Translational Research, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan, China.,Collaborative Innovation Center for Antitumor Active Substance Research and Development, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
106
|
Yuan M, Hu X, Yao L, Jiang Y, Li L. Mesenchymal stem cell homing to improve therapeutic efficacy in liver disease. Stem Cell Res Ther 2022; 13:179. [PMID: 35505419 PMCID: PMC9066724 DOI: 10.1186/s13287-022-02858-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cell (MSC) transplantation, as an alternative strategy to orthotopic liver transplantation, has been evaluated for treating end-stage liver disease. Although the therapeutic mechanism of MSC transplantation remains unclear, accumulating evidence has demonstrated that MSCs can regenerate tissues and self-renew to repair the liver through differentiation into hepatocyte-like cells, immune regulation, and anti-fibrotic mechanisms. Multiple clinical trials have confirmed that MSC transplantation restores liver function and alleviates liver damage. A sufficient number of MSCs must be home to the target tissues after administration for successful application. However, inefficient homing of MSCs after systemic administration is a major limitation in MSC therapy. Here, we review the mechanisms and clinical application status of MSCs in the treatment of liver disease and comprehensively summarize the molecular mechanisms of MSC homing, and various strategies for promoting MSC homing to improve the treatment of liver disease.
Collapse
Affiliation(s)
- Mengqin Yuan
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xue Hu
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lichao Yao
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingan Jiang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Lanjuan Li
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, China. .,State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
107
|
Riddle RB, Jennbacken K, Hansson KM, Harper MT. Endothelial inflammation and neutrophil transmigration are modulated by extracellular matrix composition in an inflammation-on-a-chip model. Sci Rep 2022; 12:6855. [PMID: 35477984 PMCID: PMC9046410 DOI: 10.1038/s41598-022-10849-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/11/2022] [Indexed: 12/20/2022] Open
Abstract
Inflammatory diseases are often characterised by excessive neutrophil infiltration from the blood stream to the site of inflammation, which damages healthy tissue and prevents resolution of inflammation. Development of anti-inflammatory drugs is hindered by lack of in vitro and in vivo models which accurately represent the disease microenvironment. In this study, we used the OrganoPlate to develop a humanized 3D in vitro inflammation-on-a-chip model to recapitulate neutrophil transmigration across the endothelium and subsequent migration through the extracellular matrix (ECM). Human umbilical vein endothelial cells formed confluent vessels against collagen I and geltrex mix, a mix of basement membrane extract and collagen I. TNF-α-stimulation of vessels upregulated inflammatory cytokine expression and promoted neutrophil transmigration. Intriguingly, major differences were found depending on the composition of the ECM. Neutrophils transmigrated in higher number and further in geltrex mix than collagen I, and did not require an N-formyl-methionyl-leucyl-phenylalanine (fMLP) gradient for transmigration. Inhibition of neutrophil proteases inhibited neutrophil transmigration on geltrex mix, but not collagen I. These findings highlight the important role of the ECM in determining cell phenotype and response to inhibitors. Future work could adapt the ECM composition for individual diseases, producing accurate models for drug development.
Collapse
Affiliation(s)
- Rebecca B Riddle
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Karin Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Kenny M Hansson
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism, R&D BioPharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
108
|
Mikucki A, McCluskey NR, Kahler CM. The Host-Pathogen Interactions and Epicellular Lifestyle of Neisseria meningitidis. Front Cell Infect Microbiol 2022; 12:862935. [PMID: 35531336 PMCID: PMC9072670 DOI: 10.3389/fcimb.2022.862935] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 02/28/2022] [Indexed: 01/17/2023] Open
Abstract
Neisseria meningitidis is a gram-negative diplococcus and a transient commensal of the human nasopharynx. It shares and competes for this niche with a number of other Neisseria species including N. lactamica, N. cinerea and N. mucosa. Unlike these other members of the genus, N. meningitidis may become invasive, crossing the epithelium of the nasopharynx and entering the bloodstream, where it rapidly proliferates causing a syndrome known as Invasive Meningococcal Disease (IMD). IMD progresses rapidly to cause septic shock and meningitis and is often fatal despite aggressive antibiotic therapy. While many of the ways in which meningococci survive in the host environment have been well studied, recent insights into the interactions between N. meningitidis and the epithelial, serum, and endothelial environments have expanded our understanding of how IMD develops. This review seeks to incorporate recent work into the established model of pathogenesis. In particular, we focus on the competition that N. meningitidis faces in the nasopharynx from other Neisseria species, and how the genetic diversity of the meningococcus contributes to the wide range of inflammatory and pathogenic potentials observed among different lineages.
Collapse
Affiliation(s)
- August Mikucki
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Nicolie R. McCluskey
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- College of Science, Health, Engineering and Education, Telethon Kids Institute, Murdoch University, Perth, WA, Australia
| | - Charlene M. Kahler
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- *Correspondence: Charlene M. Kahler,
| |
Collapse
|
109
|
Sevoflurane Dampens Acute Pulmonary Inflammation via the Adenosine Receptor A2B and Heme Oxygenase-1. Cells 2022; 11:cells11071094. [PMID: 35406657 PMCID: PMC8997763 DOI: 10.3390/cells11071094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 11/17/2022] Open
Abstract
Acute respiratory distress syndrome is a life-threatening disease associated with high mortality. The adenosine receptor A2B (Adora2b) provides anti-inflammatory effects, which are also associated with the intracellular enzyme heme oxygenase-1 (HO-1). Our study determined the mechanism of sevoflurane’s protective properties and investigated the link between sevoflurane and the impact of a functional Adora2b via HO-1 modulation during lipopolysaccharide (LPS)-induced lung injury. We examined the LPS-induced infiltration of polymorphonuclear neutrophils (PMNs) into the lung tissue and protein extravasation in wild-type and Adora2b−/− animals. We generated chimeric animals, to identify the impact of sevoflurane on Adora2b of hematopoietic and non-hematopoietic cells. Sevoflurane decreased the LPS-induced PMN-infiltration and diminished the edema formation in wild-type mice. Reduced PMN counts after sevoflurane treatment were detected only in chimeric mice, which expressed Adora2b exclusively on leukocytes. The Adora2b on hematopoietic and non-hematopoietic cells was required to improve the permeability after sevoflurane inhalation. Further, sevoflurane increased the protective effects of HO-1 modulation on PMN migration and microvascular permeability. These protective effects were abrogated by specific HO-1 inhibition. In conclusion, our data revealed new insights into the protective mechanisms of sevoflurane application during acute pulmonary inflammation and the link between sevoflurane and Adora2b, and HO-1 signaling, respectively.
Collapse
|
110
|
De Giovanni M, Tam H, Valet C, Xu Y, Looney MR, Cyster JG. GPR35 promotes neutrophil recruitment in response to serotonin metabolite 5-HIAA. Cell 2022; 185:815-830.e19. [PMID: 35148838 PMCID: PMC9037118 DOI: 10.1016/j.cell.2022.01.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/02/2022] [Accepted: 01/14/2022] [Indexed: 02/06/2023]
Abstract
Rapid neutrophil recruitment to sites of inflammation is crucial for innate immune responses. Here, we reveal that the G-protein-coupled receptor GPR35 is upregulated in activated neutrophils, and it promotes their migration. GPR35-deficient neutrophils are less recruited from blood vessels into inflamed tissue, and the mice are less efficient in clearing peritoneal bacteria. Using a bioassay, we find that serum and activated platelet supernatant stimulate GPR35, and we identify the platelet-derived serotonin metabolite 5-hydroxyindoleacetic acid (5-HIAA) as a GPR35 ligand. GPR35 function in neutrophil recruitment is strongly dependent on platelets, with the receptor promoting transmigration across platelet-coated endothelium. Mast cells also attract GPR35+ cells via 5-HIAA. Mice deficient in 5-HIAA show a loss of GPR35-mediated neutrophil recruitment to inflamed tissue. These findings identify 5-HIAA as a GPR35 ligand and neutrophil chemoattractant and establish a role for platelet- and mast cell-produced 5-HIAA in cell recruitment to the sites of inflammation and bacterial clearance.
Collapse
Affiliation(s)
- Marco De Giovanni
- Howard Hughes Medical Institute, Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Hanson Tam
- Howard Hughes Medical Institute, Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Colin Valet
- Departments of Medicine and Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ying Xu
- Howard Hughes Medical Institute, Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mark R Looney
- Departments of Medicine and Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute, Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
111
|
Dahdah A, Johnson J, Gopalkrishna S, Jaggers RM, Webb D, Murphy AJ, Hanssen NMJ, Hanaoka BY, Nagareddy PR. Neutrophil Migratory Patterns: Implications for Cardiovascular Disease. Front Cell Dev Biol 2022; 10:795784. [PMID: 35309915 PMCID: PMC8924299 DOI: 10.3389/fcell.2022.795784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/18/2022] [Indexed: 12/31/2022] Open
Abstract
The body's inflammatory response involves a series of processes that are necessary for the immune system to mitigate threats from invading pathogens. Leukocyte migration is a crucial process in both homeostatic and inflammatory states. The mechanisms involved in immune cell recruitment to the site of inflammation are numerous and require several cascades and cues of activation. Immune cells have multiple origins and can be recruited from primary and secondary lymphoid, as well as reservoir organs within the body to generate an immune response to certain stimuli. However, no matter the origin, an important aspect of any inflammatory response is the web of networks that facilitates immune cell trafficking. The vasculature is an important organ for this trafficking, especially during an inflammatory response, mainly because it allows cells to migrate towards the source of insult/injury and serves as a reservoir for leukocytes and granulocytes under steady state conditions. One of the most active and vital leukocytes in the immune system's arsenal are neutrophils. Neutrophils exist under two forms in the vasculature: a marginated pool that is attached to the vessel walls, and a demarginated pool that freely circulates within the blood stream. In this review, we seek to present the current consensus on the mechanisms involved in leukocyte margination and demargination, with a focus on the role of neutrophil migration patterns during physio-pathological conditions, in particular diabetes and cardiovascular disease.
Collapse
Affiliation(s)
- Albert Dahdah
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Jillian Johnson
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sreejit Gopalkrishna
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Robert M. Jaggers
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Darren Webb
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Andrew J. Murphy
- Division of Immunometabolism, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Nordin M. J. Hanssen
- Amsterdam Diabetes Centrum, Internal and Vascular Medicine, Amsterdam UMC, Amsterdam, Netherlands
| | - Beatriz Y. Hanaoka
- Department of Internal Medicine, Division of Rheumatology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Prabhakara R. Nagareddy
- Department of Surgery, Division of Cardiac Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
112
|
Wang L, Wei X. T Cell-Mediated Autoimmunity in Glaucoma Neurodegeneration. Front Immunol 2022; 12:803485. [PMID: 34975917 PMCID: PMC8716691 DOI: 10.3389/fimmu.2021.803485] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023] Open
Abstract
Glaucoma as the leading neurodegenerative disease leads to blindness in 3.6 million people aged 50 years and older worldwide. For many decades, glaucoma therapy has primarily focused on controlling intraocular pressure (IOP) and sound evidence supports its role in delaying the progress of retinal ganglial cell (RGC) damage and protecting patients from vision loss. Meanwhile, accumulating data point to the immune-mediated attack of the neural retina as the underlying pathological process behind glaucoma that may come independent of raised IOP. Recently, some scholars have suggested autoimmune aspects in glaucoma, with autoreactive T cells mediating the chief pathogenic process. This autoimmune process, as well as the pathological features of glaucoma, largely overlaps with other neurodegenerative diseases in the central nervous system (CNS), including Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis. In addition, immune modulation therapy, which is regarded as a potential solution for glaucoma, has been boosted in trials in some CNS neurodegenerative diseases. Thus, novel insights into the T cell-mediated immunity and treatment in CNS neurodegenerative diseases may serve as valuable inspirations for ophthalmologists. This review focuses on the role of T cell-mediated immunity in the pathogenesis of glaucoma and discusses potential applications of relevant findings of CNS neurodegenerative diseases in future glaucoma research.
Collapse
Affiliation(s)
- Lixiang Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wei
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China.,Department of Ophthalmology, Shangjin Nanfu Hospital, Chengdu, China
| |
Collapse
|
113
|
Fodil S, Arnaud M, Vaganay C, Puissant A, Lengline E, Mooney N, Itzykson R, Zafrani L. Endothelial cells: major players in acute myeloid leukaemia. Blood Rev 2022; 54:100932. [DOI: 10.1016/j.blre.2022.100932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/17/2022]
|
114
|
Mauersberger C, Hinterdobler J, Schunkert H, Kessler T, Sager HB. Where the Action Is-Leukocyte Recruitment in Atherosclerosis. Front Cardiovasc Med 2022; 8:813984. [PMID: 35087886 PMCID: PMC8787128 DOI: 10.3389/fcvm.2021.813984] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/15/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is the leading cause of death worldwide and leukocyte recruitment is a key element of this phenomenon, thus allowing immune cells to enter the arterial wall. There, in concert with accumulating lipids, the invading leukocytes trigger a plethora of inflammatory responses which promote the influx of additional leukocytes and lead to the continued growth of atherosclerotic plaques. The recruitment process follows a precise scheme of tethering, rolling, firm arrest, crawling and transmigration and involves multiple cellular and subcellular players. This review aims to provide a comprehensive up-to-date insight into the process of leukocyte recruitment relevant to atherosclerosis, each from the perspective of endothelial cells, monocytes and macrophages, neutrophils, T lymphocytes and platelets. In addition, therapeutic options targeting leukocyte recruitment into atherosclerotic lesions-or potentially arising from the growing body of insights into its precise mechanisms-are highlighted.
Collapse
Affiliation(s)
- Carina Mauersberger
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Julia Hinterdobler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Hendrik B. Sager
- Department of Cardiology, German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
115
|
Szulc-Kielbik I, Klink M. Polymorphonuclear Neutrophils and Tumors: Friend or Foe? EXPERIENTIA SUPPLEMENTUM (2012) 2022; 113:141-167. [PMID: 35165863 DOI: 10.1007/978-3-030-91311-3_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor microenvironment (TME) is a dynamic network that apart from tumor cells includes also cells of the immune system, e.g., neutrophils, which are recruited from blood circulation. In TME, neutrophils are strongly implicated in the direct and indirect interactions with tumor cells or other immune cells, and they play roles in both preventing and/or facilitating tumor progression and metastasis. The dual role of neutrophils is determined by their high plasticity and heterogeneity. Analogous to the macrophages, neutrophils can express antitumoral (N1) and protumoral (N2) phenotypes which differ substantially in morphology and function. N1 phenotype characterizes with a high cytotoxic and proinflammatory activities, while N2 phenotype with immunosuppressive and prometastatic properties. The antitumoral effect of neutrophils includes for example the production of reactive oxygen species or proapoptotic molecules. The protumoral action of neutrophils relies on releasing of proangiogenic and prometastatic mediators, immunosuppressive factors, as well as on direct helping tumor cells in extravasation process. This chapter summarizes the heterogeneity of neutrophils in TME, as well as their dual role on tumor cells.
Collapse
Affiliation(s)
| | - Magdalena Klink
- Institute of Medical Biology, Polish Academy of Sciences, Lodz, Poland.
| |
Collapse
|
116
|
Malengier-Devlies B, Metzemaekers M, Wouters C, Proost P, Matthys P. Neutrophil Homeostasis and Emergency Granulopoiesis: The Example of Systemic Juvenile Idiopathic Arthritis. Front Immunol 2021; 12:766620. [PMID: 34966386 PMCID: PMC8710701 DOI: 10.3389/fimmu.2021.766620] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/23/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are key pathogen exterminators of the innate immune system endowed with oxidative and non-oxidative defense mechanisms. More recently, a more complex role for neutrophils as decision shaping cells that instruct other leukocytes to fine-tune innate and adaptive immune responses has come into view. Under homeostatic conditions, neutrophils are short-lived cells that are continuously released from the bone marrow. Their development starts with undifferentiated hematopoietic stem cells that pass through different immature subtypes to eventually become fully equipped, mature neutrophils capable of launching fast and robust immune responses. During severe (systemic) inflammation, there is an increased need for neutrophils. The hematopoietic system rapidly adapts to this increased demand by switching from steady-state blood cell production to emergency granulopoiesis. During emergency granulopoiesis, the de novo production of neutrophils by the bone marrow and at extramedullary sites is augmented, while additional mature neutrophils are rapidly released from the marginated pools. Although neutrophils are indispensable for host protection against microorganisms, excessive activation causes tissue damage in neutrophil-rich diseases. Therefore, tight regulation of neutrophil homeostasis is imperative. In this review, we discuss the kinetics of neutrophil ontogenesis in homeostatic conditions and during emergency myelopoiesis and provide an overview of the different molecular players involved in this regulation. We substantiate this review with the example of an autoinflammatory disease, i.e. systemic juvenile idiopathic arthritis.
Collapse
Affiliation(s)
- Bert Malengier-Devlies
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mieke Metzemaekers
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Carine Wouters
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.,Division of Pediatric Rheumatology, University Hospitals Leuven, Leuven, Belgium.,European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases (RITA) at University Hospital Leuven, Leuven, Belgium
| | - Paul Proost
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Department of Microbiology, Immunology and Transplantation, Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
117
|
Lv Z, Cui B, Huang X, Feng HY, Wang T, Wang HF, Xuan YD, Li HZ, Ma X, Huang Y, Zhang X. FGL1 as a Novel Mediator and Biomarker of Malignant Progression in Clear Cell Renal Cell Carcinoma. Front Oncol 2021; 11:756843. [PMID: 34956878 PMCID: PMC8695555 DOI: 10.3389/fonc.2021.756843] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/22/2021] [Indexed: 12/18/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC), which is the most prevalent renal cell carcinoma subtype, has a poor prognosis. Emerging strategies for enhancing the immune response in ccRCC therapy are currently being investigated. Fibrinogen-like Protein 1(FGL1) is a novel mechanism that tumors may use to evade the immune system by binding LAG-3 and negatively regulating T cells. In this study, we aimed at investigating the underlying mechanism of FGL1 in ccRCC, and its expression and prognostic value. We found that FGL1 was upregulated in tumor tissues and plasma specimens of ccRCC patients. High FGL1 expression predicted a poor prognosis for ccRCC patients. We also discovered that overexpression of FGL1 enhances RCC cell migration, invasion, and metastasis by activating the epithelial-to-mesenchymal transition (EMT). Consistent with these results, we identified a significant positive correlation between expression of FGL1 and EMT-related genes through tissue microarray analysis. Gene-expression analysis revealed that FGL1-deficient ccRCC cell lines had altered transcriptional output in inflammatory response, cell-cell signaling, negative regulation of T cell activation, and intracellular signal transduction. Depletion of FGL1 significantly inhibited tumor growth and lung metastasis in orthotopic xenograft mouse model. Infiltration of myeloid-derived CD11b+ and Ly6G+ immune cells in tumor microenvironment (TME) was strikingly decreased when FGL1 expression reduced. Therefore, increased FGL1 expression in ccRCC is positively correlated with poor prognosis. Mechanistically, FGL1 facilitates the EMT process and modulates TME, which promotes ccRCC progression and metastasis. Consequently, targeting FGL1 can potentially improve clinical outcome of ccRCC patients.
Collapse
Affiliation(s)
- Zheng Lv
- School of Medicine, Nankai University, Tianjin, China
| | - Bo Cui
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xing Huang
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hua-Yi Feng
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Tao Wang
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Han-Feng Wang
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yun-Dong Xuan
- Medical School of Chinese PLA, Beijing, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hong-Zhao Li
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xin Ma
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yan Huang
- Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xu Zhang
- School of Medicine, Nankai University, Tianjin, China.,Department of Urology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
118
|
Babatunde KA, Ayuso JM, Kerr SC, Huttenlocher A, Beebe DJ. Microfluidic Systems to Study Neutrophil Forward and Reverse Migration. Front Immunol 2021; 12:781535. [PMID: 34899746 PMCID: PMC8653704 DOI: 10.3389/fimmu.2021.781535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/09/2021] [Indexed: 12/26/2022] Open
Abstract
During infection, neutrophils are the most abundantly recruited innate immune cells at sites of infection, playing critical roles in the elimination of local infection and healing of the injury. Neutrophils are considered to be short-lived effector cells that undergo cell death at infection sites and in damaged tissues. However, recent in vitro and in vivo evidence suggests that neutrophil behavior is more complex and that they can migrate away from the inflammatory site back into the vasculature following the resolution of inflammation. Microfluidic devices have contributed to an improved understanding of the interaction and behavior of neutrophils ex vivo in 2D and 3D microenvironments. The role of reverse migration and its contribution to the resolution of inflammation remains unclear. In this review, we will provide a summary of the current applications of microfluidic devices to investigate neutrophil behavior and interactions with other immune cells with a focus on forward and reverse migration in neutrophils.
Collapse
Affiliation(s)
| | - Jose M Ayuso
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, United States
| | - Sheena C Kerr
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, United States.,Carbone Cancer Center, University of Wisconsin, Madison, WI, United States
| | - Anna Huttenlocher
- Departments of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| | - David J Beebe
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI, United States.,Carbone Cancer Center, University of Wisconsin, Madison, WI, United States.,Department of Biomedical Engineering, University of Wisconsin, Madison, WI, United States
| |
Collapse
|
119
|
Neutrophil transit time and localization within the megakaryocyte define morphologically distinct forms of emperipolesis. Blood Adv 2021; 6:2081-2091. [PMID: 34872109 PMCID: PMC9006297 DOI: 10.1182/bloodadvances.2021005097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/10/2021] [Indexed: 11/30/2022] Open
Abstract
Emperipolesis (neutrophil transit through megakaryocytes) occurs in fast and slow forms that differ morphologically. Intramegakaryocytic neutrophils reside in emperisomes and in cytoplasm near the demarcation membrane system, endoplasmic reticulum, and nucleus.
Neutrophils transit through megakaryocytes in a process termed emperipolesis, but it is unknown whether this interaction is a single type of cell-in-cell interaction or a set of distinct processes. Using a murine in vitro model, we characterized emperipolesis by live-cell spinning disk microscopy and electron microscopy. Approximately half of neutrophils exited the megakaryocyte rapidly, typically in 10 minutes or less, displaying ameboid morphology as they passed through the host cell (fast emperipolesis). The remaining neutrophils assumed a sessile morphology, most remaining within the megakaryocyte for at least 60 minutes (slow emperipolesis). These neutrophils typically localized near the megakaryocyte nucleus. By ultrastructural assessment, all internalized neutrophils remained morphologically intact. Most neutrophils resided within emperisomes, but some could be visualized exiting the emperisome to enter the cell cytoplasm. Neutrophils in the cytoplasm assumed close contact with the platelet-forming demarcation membrane system or the perinuclear endoplasmic reticulum. These findings reveal that megakaryocyte emperipolesis reflects at least 2 distinct processes differing in transit time and morphology, fast and slow emperipolesis, suggesting divergent physiologic functions.
Collapse
|
120
|
Serum inter-alpha-trypsin inhibitor heavy chain 4 in patients with inflammatory bowel disease: correlation with disease risk, inflammation, activity, and its variation after treatment. Ir J Med Sci 2021; 191:2105-2111. [PMID: 34843071 DOI: 10.1007/s11845-021-02837-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/22/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4) plays vital roles in inflammatory and auto-immune diseases, but its correlations with disease risk and clinical features in inflammatory bowel disease (IBD) need further investigation. The present study intended to explore the correlation of ITIH4 with disease activity and inflammation, as well as its change after treatment in IBD patients. METHODS Totally, 40 active Crohn's disease (A-CD) patients, 40 clinical-remission CD (R-CD) patients, 40 active ulcerative colitis (A-UC) patients, 40 clinical-remission UC (R-UC) patients, and 40 health controls (HCs) were enrolled. ITIH4 in serum was assessed by ELISA. RESULTS ITIH4 was lower in A-CD, R-CD, A-UC, and R-UC patients than in HCs (P < 0.001). Notably, ITIH4 reduced in A-CD patients than in R-CD patients (P = 0.017), and in A-UC patients compared with R-UC patients (P = 0.010). Besides, in A-CD patients, ITIH4 negatively correlated with tumor necrosis factor-alpha (TNF-α), interleukin (IL)-17A, IL-1β, C-reactive protein (CRP), and clinical disease activity index score (all P < 0.05). In A-UC patients, ITIH4 negatively correlated with TNF-α, IL-17A, IL-1β, IL-6, CRP, and Mayo score (all P < 0.05). However, in R-CD and R-UC patients, these correlations were less obvious than in A-CD and A-UC patients. ITIH4 was increased after treatment (all P < 0.05), and its expression at W12 after treatment was higher in response patients compared with no response patients in A-CD (P = 0.022) and A-UC groups (P = 0.038). CONCLUSION ITIH4 correlates with IBD susceptibility, active risk, inflammation level, and its elevation after treatment relates to clinical response in IBD patients.
Collapse
|
121
|
Wang H, Lv G, Lian S, Wang J, Wu R. Effect of Copper, Zinc, and Selenium on the Migration of Bovine Neutrophils. Vet Sci 2021; 8:vetsci8110281. [PMID: 34822654 PMCID: PMC8618487 DOI: 10.3390/vetsci8110281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 01/15/2023] Open
Abstract
Neutrophils represent the first line of mammary gland defense against invading pathogens by transmigration across the mammary epithelial cell barrier. The effect of trace elements on the migration of bovine neutrophils is not clear. In this study, we investigated the effect of copper (Cu; 0.5, 1.0 and 1.5 mg/L), zinc (Zn; 1.0, 5.0 and 10 mg/L) and selenium (Se; 0.1, 1.0 and 2.0 mg/L) on the migration of bovine neutrophils by using a Transwell assay. The results showed that Cu, Zn and Se promoted the number of neutrophils in the trans-mammary epithelium. With the increased concentration of Cu at 1.5 mg/L, the number of neutrophils in the trans-mammary epithelium was increased significantly (p < 0.05). Zn (5.0 mg/L) and Se (0.1 mg/L) increased the migrated number of neutrophils (p < 0.01) to an extremely significant degree. These findings provided a theoretical and experimental basis for mammary gland immunity in dairy cows. Thus, we suggest that adding moderate amounts of different trace elements can improve the immune function of dairy cows.
Collapse
Affiliation(s)
- Hai Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (H.W.); (G.L.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Guanxin Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (H.W.); (G.L.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (H.W.); (G.L.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (H.W.); (G.L.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
| | - Rui Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China; (H.W.); (G.L.); (S.L.); (J.W.)
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing 163319, China
- Correspondence: ; Tel.: +86-459-6819188
| |
Collapse
|
122
|
Oliveira-Costa KM, Menezes GB, Paula Neto HA. Neutrophil accumulation within tissues: A damage x healing dichotomy. Biomed Pharmacother 2021; 145:112422. [PMID: 34781139 DOI: 10.1016/j.biopha.2021.112422] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 02/09/2023] Open
Abstract
The abundance of neutrophils in human circulation, their fast mobilization from blood to tissues, along with their alleged short life-span led to the image of neutrophils as a homogeneous cell type designed to fight infections and die in the process. Additionally, their granule content and capacity to produce molecules with considerable cytotoxic potential, lead to the general belief that neutrophil activation inexorably results in side effect of extensive tissue injury. Neutrophil activation in fact causes tissue injury as an adverse effect, but it seems that this is restricted to particular pathological situations and more of an "exception to the rule". Here we review evidences arising especially from intravital microscopy studies that demonstrate neutrophils as cells endowed with sophisticated mechanisms and able to engage in complex interactions as to minimize damage and optimize their effector functions. Moreover, neutrophil infiltration may even contribute to tissue healing and repair which may altogether demand a reexamination of current anti-inflammatory therapies that have neutrophil migration and activation as a target.
Collapse
Affiliation(s)
- Karen Marques Oliveira-Costa
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Gustavo B Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Heitor A Paula Neto
- Laboratório de Alvos Moleculares, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
123
|
Sionov RV. Leveling Up the Controversial Role of Neutrophils in Cancer: When the Complexity Becomes Entangled. Cells 2021; 10:cells10092486. [PMID: 34572138 PMCID: PMC8465406 DOI: 10.3390/cells10092486] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are the most abundant immune cell in the circulation of human and act as gatekeepers to discard foreign elements that have entered the body. They are essential in initiating immune responses for eliminating invaders, such as microorganisms and alien particles, as well as to act as immune surveyors of cancer cells, especially during the initial stages of carcinogenesis and for eliminating single metastatic cells in the circulation and in the premetastatic organs. Since neutrophils can secrete a whole range of factors stored in their many granules as well as produce reactive oxygen and nitrogen species upon stimulation, neutrophils may directly or indirectly affect carcinogenesis in both the positive and negative directions. An intricate crosstalk between tumor cells, neutrophils, other immune cells and stromal cells in the microenvironment modulates neutrophil function resulting in both anti- and pro-tumor activities. Both the anti-tumor and pro-tumor activities require chemoattraction towards the tumor cells, neutrophil activation and ROS production. Divergence is seen in other neutrophil properties, including differential secretory repertoire and membrane receptor display. Many of the direct effects of neutrophils on tumor growth and metastases are dependent on tight neutrophil–tumor cell interactions. Among them, the neutrophil Mac-1 interaction with tumor ICAM-1 and the neutrophil L-selectin interaction with tumor-cell sialomucins were found to be involved in the neutrophil-mediated capturing of circulating tumor cells resulting in increased metastatic seeding. On the other hand, the anti-tumor function of neutrophils was found to rely on the interaction between tumor-surface-expressed receptor for advanced glycation end products (RAGE) and Cathepsin G expressed on the neutrophil surface. Intriguingly, these two molecules are also involved in the promotion of tumor growth and metastases. RAGE is upregulated during early inflammation-induced carcinogenesis and was found to be important for sustaining tumor growth and homing at metastatic sites. Cathepsin G was found to be essential for neutrophil-supported lung colonization of cancer cells. These data level up the complexity of the dual role of neutrophils in cancer.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- Hadassah Medical School, The Hebrew University of Jerusalem, Ein Kerem Campus, P.O.B. 12272, Jerusalem 9112102, Israel
| |
Collapse
|
124
|
Salminen AT, Tithof J, Izhiman Y, Masters EA, McCloskey MC, Gaborski TR, Kelley DH, Pietropaoli AP, Waugh RE, McGrath JL. Endothelial cell apicobasal polarity coordinates distinct responses to luminally versus abluminally delivered TNF-α in a microvascular mimetic. Integr Biol (Camb) 2021; 12:275-289. [PMID: 33164044 DOI: 10.1093/intbio/zyaa022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/28/2020] [Accepted: 10/05/2020] [Indexed: 12/27/2022]
Abstract
Endothelial cells (ECs) are an active component of the immune system and interact directly with inflammatory cytokines. While ECs are known to be polarized cells, the potential role of apicobasal polarity in response to inflammatory mediators has been scarcely studied. Acute inflammation is vital in maintaining healthy tissue in response to infection; however, chronic inflammation can lead to the production of systemic inflammatory cytokines and deregulated leukocyte trafficking, even in the absence of a local infection. Elevated levels of cytokines in circulation underlie the pathogenesis of sepsis, the leading cause of intensive care death. Because ECs constitute a key barrier between circulation (luminal interface) and tissue (abluminal interface), we hypothesize that ECs respond differentially to inflammatory challenge originating in the tissue versus circulation as in local and systemic inflammation, respectively. To begin this investigation, we stimulated ECs abluminally and luminally with the inflammatory cytokine tumor necrosis factor alpha (TNF-α) to mimic a key feature of local and systemic inflammation, respectively, in a microvascular mimetic (μSiM-MVM). Polarized IL-8 secretion and polymorphonuclear neutrophil (PMN) transmigration were quantified to characterize the EC response to luminal versus abluminal TNF-α. We observed that ECs uniformly secrete IL-8 in response to abluminal TNF-α and is followed by PMN transmigration. The response to abluminal treatment was coupled with the formation of ICAM-1-rich membrane ruffles on the apical surface of ECs. In contrast, luminally stimulated ECs secreted five times more IL-8 into the luminal compartment than the abluminal compartment and sequestered PMNs on the apical EC surface. Our results identify clear differences in the response of ECs to TNF-α originating from the abluminal versus luminal side of a monolayer for the first time and may provide novel insight into future inflammatory disease intervention strategies.
Collapse
Affiliation(s)
- Alec T Salminen
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Jeffrey Tithof
- Department of Mechanical Engineering, University of Rochester, Rochester, NY, USA
| | - Yara Izhiman
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Elysia A Masters
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Molly C McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Thomas R Gaborski
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, USA
| | - Douglas H Kelley
- Department of Mechanical Engineering, University of Rochester, Rochester, NY, USA
| | - Anthony P Pietropaoli
- Medicine, Pulmonary Disease and Critical Care, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard E Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| |
Collapse
|
125
|
Small extracellular vesicle-mediated bidirectional crosstalk between neutrophils and tumor cells. Cytokine Growth Factor Rev 2021; 61:16-26. [PMID: 34479816 DOI: 10.1016/j.cytogfr.2021.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023]
Abstract
Neutrophils are the first line of defense against tissue injury and play an important role in tumor progression. Tumor-associated neutrophils (TANs) mediate pro-tumor immunosuppressive activity and their infiltration into tumors is associated with poor outcome in a variety of malignant diseases. The tumor cell-neutrophil crosstalk is mediated by small extracellular vesicles (sEVs) also referred to as exosomes which represent a major mechanism for intercellular communication. This review will address the role of neutrophil-derived sEVs (NEX) in reprogramming the TME and on mechanisms that regulate the dual potential of NEX to promote tumor progression on one hand and suppress tumor growth on the other. Emerging data suggest that both, NEX and tumor-derived sEVs (TEX) carry complex molecular cargos which upon delivery to recipient cells in the tumor microenvironment (TME) modulate their behavior and reprogram them to mediate pro-inflammatory or immunosuppressive responses. Although it remains unknown how the balance between the often conflicting signaling of TEX and NEX is regulated, this review is an attempt to provide insights into mechanisms that underpin this complex bidirectional crosstalk. A better understanding of the signals NEX process or deliver in the TME might lead to the development of novel approaches to the control of tumor progression in the future.
Collapse
|
126
|
Pleskova SN, Bobyk SZ, Kriukov RN, Gorshkova EN, Novikov DV, Vasilchikov PI, Bezrukov NA, Novikov VV. S. aureus and E. coli change the force and work of adhesion between P- and E-selectins of endothelial cells and ligands of neutrophil granulocytes. Micron 2021; 150:103139. [PMID: 34428610 DOI: 10.1016/j.micron.2021.103139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/14/2021] [Accepted: 08/18/2021] [Indexed: 01/10/2023]
Abstract
Thanks to the modification of the force spectroscopy method, when a neutrophil is fixed on the tip, and an endotheliocyte culture is grown on the substrate, the exact indicators of the adhesion force and adhesion work between cells have been investigated. The high variability of adhesion contacts in different donors associated with different expression profiles of neutrophils. It was found by flow cytometry that the EA.hy926 cell line actively expresses VCAM-1, as well as P- and E-selectin under the Staphylococcus aureus influence after 60 min of co-incubation. At the same time, the integral indicators of the adhesion force and adhesion work in the "neutrophil - endothelial cell" interaction were significantly inhibited by S. aureus in all studied donors. Since the VCAM-1 receptor is not involved in the adhesion bonds between neutrophils and endothelial cells, the suppression of the interaction is associated with the inhibition of P- and E-selectins, but direct receptors removal from the endothelial cells surface of the EA.hy926 cell line does not occur. Escherichia coli causes multidirectional effects in the system of interaction "neutrophil - endothelial cell", depending on the expression profile of the donor's neutrophils. However, the cumulative effect of interaction from all donors shows that in general, under the influence of E. coli, there is an increase in adhesion force and a suppression of adhesion work.
Collapse
Affiliation(s)
- S N Pleskova
- Research and Education Center for Physics of Solid State Nanostructures, Lobachevsky State University of Nizhny Novgorod, Gagarina Ave., 23, 603950, Nizhny Novgorod, Russia; Department "Nanotechnology and Biotechnology", R.E. Alekseev Technical State University of Nizhny Novgorod, Minina St., 24, 603155, Nizhny Novgorod, Russia.
| | - S Z Bobyk
- Research and Education Center for Physics of Solid State Nanostructures, Lobachevsky State University of Nizhny Novgorod, Gagarina Ave., 23, 603950, Nizhny Novgorod, Russia
| | - R N Kriukov
- Research and Education Center for Physics of Solid State Nanostructures, Lobachevsky State University of Nizhny Novgorod, Gagarina Ave., 23, 603950, Nizhny Novgorod, Russia
| | - E N Gorshkova
- Department of Molecular Biology and Immunology, Lobachevsky State University of Nizhny Novgorod, Gagarina Ave., 23, 603950, Nizhny Novgorod, Russia
| | - D V Novikov
- Nizhniy Novgorod Research Institute of Epidemiology and Microbiology named after Academician I.N. Blokhina, Malaya Yamskaya St., 71, Nizhny Novgorod, Russia
| | - P I Vasilchikov
- Department of Molecular Biology and Immunology, Lobachevsky State University of Nizhny Novgorod, Gagarina Ave., 23, 603950, Nizhny Novgorod, Russia
| | - N A Bezrukov
- Department "Nanotechnology and Biotechnology", R.E. Alekseev Technical State University of Nizhny Novgorod, Minina St., 24, 603155, Nizhny Novgorod, Russia
| | - V V Novikov
- Nizhniy Novgorod Research Institute of Epidemiology and Microbiology named after Academician I.N. Blokhina, Malaya Yamskaya St., 71, Nizhny Novgorod, Russia
| |
Collapse
|
127
|
Liu X, Shi GP, Guo J. Innate Immune Cells in Pressure Overload-Induced Cardiac Hypertrophy and Remodeling. Front Cell Dev Biol 2021; 9:659666. [PMID: 34368120 PMCID: PMC8343105 DOI: 10.3389/fcell.2021.659666] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/28/2021] [Indexed: 12/23/2022] Open
Abstract
Pressure overload and heart failure are among the leading causes of cardiovascular morbidity and mortality. Accumulating evidence suggests that inflammatory cell activation and release of inflammatory mediators are of vital importance during the pathogenesis of these cardiac diseases. Yet, the roles of innate immune cells and subsequent inflammatory events in these processes remain poorly understood. Here, we outline the possible underlying mechanisms of innate immune cell participation, including mast cells, macrophages, monocytes, neutrophils, dendritic cells, eosinophils, and natural killer T cells in these pathological processes. Although these cells accumulate in the atrium or ventricles at different time points after pressure overload, their cardioprotective or cardiodestructive activities differ from each other. Among them, mast cells, neutrophils, and dendritic cells exert detrimental function in experimental models, whereas eosinophils and natural killer T cells display cardioprotective activities. Depending on their subsets, macrophages and monocytes may exacerbate cardiodysfunction or negatively regulate cardiac hypertrophy and remodeling. Pressure overload stimulates the secretion of cytokines, chemokines, and growth factors from innate immune cells and even resident cardiomyocytes that together assist innate immune cell infiltration into injured heart. These infiltrates are involved in pro-hypertrophic events and cardiac fibroblast activation. Immune regulation of cardiac innate immune cells becomes a promising therapeutic approach in experimental cardiac disease treatment, highlighting the significance of their clinical evaluation in humans.
Collapse
Affiliation(s)
- Xin Liu
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Junli Guo
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research & Key Laboratory of Emergency and Trauma of Ministry of Education, Institute of Cardiovascular Research of the First Affiliated Hospital, Hainan Medical University, Haikou, China
| |
Collapse
|
128
|
Kinoshita M, Ogawa Y, Hama N, Ujiie I, Hasegawa A, Nakajima S, Nomura T, Adachi J, Sato T, Koizumi S, Shimada S, Fujita Y, Takahashi H, Mizukawa Y, Tomonaga T, Nagao K, Abe R, Kawamura T. Neutrophils initiate and exacerbate Stevens-Johnson syndrome and toxic epidermal necrolysis. Sci Transl Med 2021; 13:13/600/eaax2398. [PMID: 34193610 DOI: 10.1126/scitranslmed.aax2398] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 12/13/2020] [Accepted: 06/10/2021] [Indexed: 12/14/2022]
Abstract
Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are life-threatening mucocutaneous adverse drug reactions characterized by massive epidermal detachment. Cytotoxic T cells and associated effector molecules are known to drive SJS/TEN pathophysiology, but the contribution of innate immune responses is not well understood. We describe a mechanism by which neutrophils triggered inflammation during early phases of SJS/TEN. Skin-infiltrating CD8+ T cells produced lipocalin-2 in a drug-specific manner, which triggered the formation of neutrophil extracellular traps (NETs) in early lesional skin. Neutrophils undergoing NETosis released LL-37, an antimicrobial peptide, which induced formyl peptide receptor 1 (FPR1) expression by keratinocytes. FPR1 expression caused keratinocytes to be vulnerable to necroptosis that caused further release of LL-37 by necroptotic keratinocytes and induced FPR1 expression on surrounding keratinocytes, which likely amplified the necroptotic response. The NETs-necroptosis axis was not observed in less severe cutaneous adverse drug reactions, autoimmune diseases, or neutrophil-associated disorders, suggesting that this was a process specific to SJS/TEN. Initiation and progression of SJS/TEN keratinocyte necroptosis appear to involve a cascade of events mediated by innate and adaptive immune responses, and understanding these responses may contribute to the identification of diagnostic markers or therapeutic targets for these adverse drug reactions.
Collapse
Affiliation(s)
- Manao Kinoshita
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Youichi Ogawa
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Natsumi Hama
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Inkin Ujiie
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Akito Hasegawa
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Nomura
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Jun Adachi
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Takuya Sato
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shinji Shimada
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yasuyuki Fujita
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hayato Takahashi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiko Mizukawa
- Department of Dermatology, Kyorin University School of Medicine, Tokyo, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Proteomics for Drug Discovery, Center for Drug Design Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Keisuke Nagao
- Cutaneous Leukocyte Biology Section, Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health, Bethesda, MD, USA
| | - Riichiro Abe
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Tatsuyoshi Kawamura
- Department of Dermatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
129
|
Platelet and Erythrocyte Extravasation across Inflamed Corneal Venules Depend on CD18, Neutrophils, and Mast Cell Degranulation. Int J Mol Sci 2021; 22:ijms22147360. [PMID: 34298979 PMCID: PMC8329926 DOI: 10.3390/ijms22147360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 01/26/2023] Open
Abstract
Platelet extravasation during inflammation is under-appreciated. In wild-type (WT) mice, a central corneal epithelial abrasion initiates neutrophil (PMN) and platelet extravasation from peripheral limbal venules. The same injury in mice expressing low levels of the β2-integrin, CD18 (CD18hypo mice) shows reduced platelet extravasation with PMN extravasation apparently unaffected. To better define the role of CD18 on platelet extravasation, we focused on two relevant cell types expressing CD18: PMNs and mast cells. Following corneal abrasion in WT mice, we observed not only extravasated PMNs and platelets but also extravasated erythrocytes (RBCs). Ultrastructural observations of engorged limbal venules showed platelets and RBCs passing through endothelial pores. In contrast, injured CD18hypo mice showed significantly less venule engorgement and markedly reduced platelet and RBC extravasation; mast cell degranulation was also reduced compared to WT mice. Corneal abrasion in mast cell-deficient (KitW-sh/W-sh) mice showed less venule engorgement, delayed PMN extravasation, reduced platelet and RBC extravasation and delayed wound healing compared to WT mice. Finally, antibody-induced depletion of circulating PMNs prior to corneal abrasion reduced mast cell degranulation, venule engorgement, and extravasation of PMNs, platelets, and RBCs. In summary, in the injured cornea, platelet and RBC extravasation depends on CD18, PMNs, and mast cell degranulation.
Collapse
|
130
|
HIF2α is a direct regulator of neutrophil motility. Blood 2021; 137:3416-3427. [PMID: 33619535 DOI: 10.1182/blood.2020007505] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 01/24/2021] [Indexed: 12/19/2022] Open
Abstract
Orchestrated recruitment of neutrophils to inflamed tissue is essential during the initiation of inflammation. Inflamed areas are usually hypoxic, and adaptation to reduced oxygen pressure is typically mediated by hypoxia pathway proteins. However, it remains unclear how these factors influence the migration of neutrophils to and at the site of inflammation during their transmigration through the blood-endothelial cell barrier, as well as their motility in the interstitial space. Here, we reveal that activation of hypoxia-inducible factor 2 (HIF2α) as a result of a deficiency in HIF prolyl hydroxylase domain protein 2 (PHD2) boosts neutrophil migration specifically through highly confined microenvironments. In vivo, the increased migratory capacity of PHD2-deficient neutrophils resulted in massive tissue accumulation in models of acute local inflammation. Using systematic RNA sequencing analyses and mechanistic approaches, we identified RhoA, a cytoskeleton organizer, as the central downstream factor that mediates HIF2α-dependent neutrophil motility. Thus, we propose that the novel PHD2-HIF2α-RhoA axis is vital to the initial stages of inflammation because it promotes neutrophil movement through highly confined tissue landscapes.
Collapse
|
131
|
Endothelial Dysfunction and Neutrophil Degranulation as Central Events in Sepsis Physiopathology. Int J Mol Sci 2021; 22:ijms22126272. [PMID: 34200950 PMCID: PMC8230689 DOI: 10.3390/ijms22126272] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a major health problem worldwide. It is a time-dependent disease, with a high rate of morbidity and mortality. In this sense, an early diagnosis is essential to reduce these rates. The progressive increase of both the incidence and prevalence of sepsis has translated into a significant socioeconomic burden for health systems. Currently, it is the leading cause of noncoronary mortality worldwide and represents one of the most prevalent pathologies both in hospital emergency services and in intensive care units. In this article, we review the role of both endothelial dysfunction and neutrophil dysregulation in the physiopathology of this disease. The lack of a key symptom in sepsis makes it difficult to obtain a quick and accurate diagnosis of this condition. Thus, it is essential to have fast and reliable diagnostic tools. In this sense, the use of biomarkers can be a very important alternative when it comes to achieving these goals. Both new biomarkers and treatments related to endothelial dysfunction and neutrophil dysregulation deserve to be further investigated in order to open new venues for the diagnosis, treatment and prognosis of sepsis.
Collapse
|
132
|
Richardson IM, Calo CJ, Hind LE. Microphysiological Systems for Studying Cellular Crosstalk During the Neutrophil Response to Infection. Front Immunol 2021; 12:661537. [PMID: 33986752 PMCID: PMC8111168 DOI: 10.3389/fimmu.2021.661537] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are the primary responders to infection, rapidly migrating to sites of inflammation and clearing pathogens through a variety of antimicrobial functions. This response is controlled by a complex network of signals produced by vascular cells, tissue resident cells, other immune cells, and the pathogen itself. Despite significant efforts to understand how these signals are integrated into the neutrophil response, we still do not have a complete picture of the mechanisms regulating this process. This is in part due to the inherent disadvantages of the most-used experimental systems: in vitro systems lack the complexity of the tissue microenvironment and animal models do not accurately capture the human immune response. Advanced microfluidic devices incorporating relevant tissue architectures, cell-cell interactions, and live pathogen sources have been developed to overcome these challenges. In this review, we will discuss the in vitro models currently being used to study the neutrophil response to infection, specifically in the context of cell-cell interactions, and provide an overview of their findings. We will also provide recommendations for the future direction of the field and what important aspects of the infectious microenvironment are missing from the current models.
Collapse
Affiliation(s)
| | | | - Laurel E. Hind
- Department of Chemical and Biological Engineering, University of Colorado – Boulder, Boulder, CO, United States
| |
Collapse
|
133
|
Rébillard RM, Charabati M, Grasmuck C, Filali-Mouhim A, Tastet O, Brassard N, Daigneault A, Bourbonnière L, Anand SP, Balthazard R, Beaudoin-Bussières G, Gasser R, Benlarbi M, Moratalla AC, Solorio YC, Boutin M, Farzam-Kia N, Descôteaux-Dinelle J, Fournier AP, Gowing E, Laumaea A, Jamann H, Lahav B, Goyette G, Lemaître F, Mamane VH, Prévost J, Richard J, Thai K, Cailhier JF, Chomont N, Finzi A, Chassé M, Durand M, Arbour N, Kaufmann DE, Prat A, Larochelle C. Identification of SARS-CoV-2-specific immune alterations in acutely ill patients. J Clin Invest 2021; 131:145853. [PMID: 33635833 DOI: 10.1172/jci145853] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/19/2021] [Indexed: 01/08/2023] Open
Abstract
Dysregulated immune profiles have been described in symptomatic patients infected with SARS-CoV-2. Whether the reported immune alterations are specific to SARS-CoV-2 infection or also triggered by other acute illnesses remains unclear. We performed flow cytometry analysis on fresh peripheral blood from a consecutive cohort of (a) patients hospitalized with acute SARS-CoV-2 infection, (b) patients of comparable age and sex hospitalized for another acute disease (SARS-CoV-2 negative), and (c) healthy controls. Using both data-driven and hypothesis-driven analyses, we found several dysregulations in immune cell subsets (e.g., decreased proportion of T cells) that were similarly associated with acute SARS-CoV-2 infection and non-COVID-19-related acute illnesses. In contrast, we identified specific differences in myeloid and lymphocyte subsets that were associated with SARS-CoV-2 status (e.g., elevated proportion of ICAM-1+ mature/activated neutrophils, ALCAM+ monocytes, and CD38+CD8+ T cells). A subset of SARS-CoV-2-specific immune alterations correlated with disease severity, disease outcome at 30 days, and mortality. Our data provide an understanding of the immune dysregulation specifically associated with SARS-CoV-2 infection among acute care hospitalized patients. Our study lays the foundation for the development of specific biomarkers to stratify SARS-CoV-2-positive patients at risk of unfavorable outcomes and to uncover candidate molecules to investigate from a therapeutic perspective.
Collapse
Affiliation(s)
- Rose-Marie Rébillard
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marc Charabati
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Camille Grasmuck
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Abdelali Filali-Mouhim
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Olivier Tastet
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Nathalie Brassard
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Audrey Daigneault
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Lyne Bourbonnière
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Sai Priya Anand
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Renaud Balthazard
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Guillaume Beaudoin-Bussières
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Romain Gasser
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Mehdi Benlarbi
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Ana Carmena Moratalla
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Yves Carpentier Solorio
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marianne Boutin
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Negar Farzam-Kia
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Jade Descôteaux-Dinelle
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Antoine Philippe Fournier
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Elizabeth Gowing
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Annemarie Laumaea
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Hélène Jamann
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Boaz Lahav
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Guillaume Goyette
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Florent Lemaître
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Victoria Hannah Mamane
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Jérémie Prévost
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Jonathan Richard
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Karine Thai
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Jean-François Cailhier
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Nicolas Chomont
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Andrés Finzi
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada.,Department of Microbiology, Infectious Diseases and Immunology, and
| | - Michaël Chassé
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Madeleine Durand
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Nathalie Arbour
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Daniel E Kaufmann
- Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Catherine Larochelle
- Department of Neuroscience, Université de Montréal, Montreal, Quebec, Canada.,Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| |
Collapse
|
134
|
Blanch-Ruiz MA, Ortega-Luna R, Martínez-Cuesta MÁ, Álvarez Á. The Neutrophil Secretome as a Crucial Link between Inflammation and Thrombosis. Int J Mol Sci 2021; 22:4170. [PMID: 33920656 PMCID: PMC8073391 DOI: 10.3390/ijms22084170] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases are a leading cause of death. Blood-cell interactions and endothelial dysfunction are fundamental in thrombus formation, and so further knowledge of the pathways involved in such cellular crosstalk could lead to new therapeutical approaches. Neutrophils are secretory cells that release well-known soluble inflammatory signaling mediators and other complex cellular structures whose role is not fully understood. Studies have reported that neutrophil extracellular vesicles (EVs) and neutrophil extracellular traps (NETs) contribute to thrombosis. The objective of this review is to study the role of EVs and NETs as key factors in the transition from inflammation to thrombosis. The neutrophil secretome can promote thrombosis due to the presence of different factors in the EVs bilayer that can trigger blood clotting, and to the release of soluble mediators that induce platelet activation or aggregation. On the other hand, one of the main pathways by which NETs induce thrombosis is through the creation of a scaffold to which platelets and other blood cells adhere. In this context, platelet activation has been associated with the induction of NETs release. Hence, the structure and composition of EVs and NETs, as well as the feedback mechanism between the two processes that causes pathological thrombus formation, require exhaustive analysis to clarify their role in thrombosis.
Collapse
Affiliation(s)
- María Amparo Blanch-Ruiz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - María Ángeles Martínez-Cuesta
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| |
Collapse
|
135
|
Jahan D, Peile E, Sheikh MA, Islam S, Parasnath S, Sharma P, Iskandar K, Dhingra S, Charan J, Hardcastle TC, Samad N, Chowdhury TS, Dutta S, Haque M. Is it time to reconsider prophylactic antimicrobial use for hematopoietic stem cell transplantation? a narrative review of antimicrobials in stem cell transplantation. Expert Rev Anti Infect Ther 2021; 19:1259-1280. [PMID: 33711240 DOI: 10.1080/14787210.2021.1902304] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Hematopoietic Stem Cell Transplantation (HSCT) is a life-saving procedure for multiple types of hematological cancer, autoimmune diseases, and genetic-linked metabolic diseases in humans. Recipients of HSCT transplant are at high risk of microbial infections that significantly correlate with the presence of graft-versus-host disease (GVHD) and the degree of immunosuppression. Infection in HSCT patients is a leading cause of life-threatening complications and mortality. AREAS COVERED This review covers issues pertinent to infection in the HSCT patient, including bacterial and viral infection; strategies to reduce GVHD; infection patterns; resistance and treatment options; adverse drug reactions to antimicrobials, problems of antimicrobial resistance; perturbation of the microbiome; the role of prebiotics, probiotics, and antimicrobial peptides. We highlight potential strategies to minimize the use of antimicrobials. EXPERT OPINION Measures to control infection and its transmission remain significant HSCT management policy and planning issues. Transplant centers need to consider carefully prophylactic use of antimicrobials for neutropenic patients. The judicious use of appropriate antimicrobials remains a crucial part of the treatment protocol. However, antimicrobials' adverse effects cause microbiome diversity and dysbiosis and have been shown to increase morbidity and mortality.
Collapse
Affiliation(s)
- Dilshad Jahan
- Department of Hematology, Asgar Ali Hospital, 111/1/A Distillery Road, Gandaria Beside Dhupkhola, Dhaka 1204, Bangladesh
| | - Ed Peile
- Department of Medical Education, Warwick Medical School, University of Warwick, Coventry, UK
| | | | - Salequl Islam
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Sharlene Parasnath
- Department of Clinical Hematology, Inkosi Albert Luthuli Central Hospital, 800 Vusi Mzimela Road, Cato Manor, Durban, South Africa
| | - Paras Sharma
- Department of Pharmacognosy, BVM College of Pharmacy, Gwalior, India
| | - Katia Iskandar
- Lebanese University, School of Pharmacy, Beirut, Lebanon.,INSPECT-LB: Institute National de Sante Publique, Epidemiologie Clinique et Toxicologie, Beirut, Lebanon.,Universite Paul Sabatier UT3, INSERM, UMR1027, Toulouse, France
| | - Sameer Dhingra
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar, India
| | - Jaykaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Timothy Craig Hardcastle
- Trauma Service, Inkosi Albert Luthuli Central Hospital, Mayville, South Africa.,Department of Surgery, Nelson R Mandela School of Clinical Medicine, UKZN, South Africa
| | - Nandeeta Samad
- Department of Public Health, North South University, Bangladesh
| | | | - Siddhartha Dutta
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Mainul Haque
- The Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sungai Besi, Kuala Lumpur, Malaysia
| |
Collapse
|
136
|
Sekheri M, Othman A, Filep JG. β2 Integrin Regulation of Neutrophil Functional Plasticity and Fate in the Resolution of Inflammation. Front Immunol 2021; 12:660760. [PMID: 33859651 PMCID: PMC8043047 DOI: 10.3389/fimmu.2021.660760] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 03/08/2021] [Indexed: 12/31/2022] Open
Abstract
Neutrophils act as the first line of cellular defense against invading pathogens or tissue injury. Their rapid recruitment into inflamed tissues is critical for the elimination of invading microorganisms and tissue repair, but is also capable of inflicting damage to neighboring tissues. The β2 integrins and Mac-1 (CD11b/CD18, αMβ2 or complement receptor 3) in particular, are best known for mediating neutrophil adhesion and transmigration across the endothelium and phagocytosis of microbes. However, Mac-1 has a broad ligand recognition property that contributes to the functional versatility of the neutrophil population far beyond their antimicrobial function. Accumulating evidence over the past decade has demonstrated roles for Mac-1 ligands in regulating reverse neutrophil transmigration, lifespan, phagocytosis-induced cell death, release of neutrophil extracellular traps and efferocytosis, hence extending the traditional β2 integrin repertoire in shaping innate and adaptive immune responses. Understanding the functions of β2 integrins may partly explain neutrophil heterogeneity and may be instrumental to develop novel therapies specifically targeting Mac-1-mediated pro-resolution actions without compromising immunity. Thus, this review details novel insights on outside-in signaling through β2 integrins and neutrophil functional heterogeneity pertinent to the resolution of inflammation.
Collapse
Affiliation(s)
- Meriem Sekheri
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada.,Department of Biomedical Sciences, University of Montreal, Montreal, QC, Canada.,Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| | - Amira Othman
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada.,Department of Biomedical Sciences, University of Montreal, Montreal, QC, Canada.,Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada.,Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| |
Collapse
|
137
|
Guo K, Ma S. The Immune System in Transfusion-Related Acute Lung Injury Prevention and Therapy: Update and Perspective. Front Mol Biosci 2021; 8:639976. [PMID: 33842545 PMCID: PMC8024523 DOI: 10.3389/fmolb.2021.639976] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
As an initiator of respiratory distress, transfusion-related acute lung injury (TRALI) is regarded as one of the rare complications associated with transfusion medicine. However, to date, the pathogenesis of TRALI is still unclear, and specific therapies are unavailable. Understanding the mechanisms of TRALI may promote the design of preventive and therapeutic strategies. The immune system plays vital roles in reproduction, development and homeostasis. Sterile tissue damage, such as physical trauma, ischemia, or reperfusion injury, induces an inflammatory reaction that results in wound healing and regenerative mechanisms. In other words, in addition to protecting against pathogens, the immune response may be strongly associated with TRALI prevention and treatment through a variety of immunomodulatory strategies to inhibit excessive immune system activation. Immunotherapy based on immune cells or immunological targets may eradicate complications. For example, IL-10 therapy is a promising therapeutic strategy to explore further. This review will focus on ultramodern advances in our understanding of the potential role of the immune system in TRALI prevention and treatment.
Collapse
Affiliation(s)
- Kai Guo
- Department of Transfusion Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Shuxuan Ma
- Department of Transfusion Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
138
|
Boric MP, Durán WN, Figueroa XF. Editorial: Cell Communication in Vascular Biology. Front Physiol 2021; 12:656959. [PMID: 33746785 PMCID: PMC7965979 DOI: 10.3389/fphys.2021.656959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/09/2021] [Indexed: 11/18/2022] Open
Affiliation(s)
- Mauricio P Boric
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Walter N Durán
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Xavier F Figueroa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
139
|
Othman A, Sekheri M, Filep JG. Roles of neutrophil granule proteins in orchestrating inflammation and immunity. FEBS J 2021; 289:3932-3953. [PMID: 33683814 PMCID: PMC9546106 DOI: 10.1111/febs.15803] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/17/2021] [Accepted: 03/05/2021] [Indexed: 12/17/2022]
Abstract
Neutrophil granulocytes form the first line of host defense against invading pathogens and tissue injury. They are rapidly recruited from the blood to the affected sites, where they deploy an impressive arsenal of effectors to eliminate invading microbes and damaged cells. This capacity is endowed in part by readily mobilizable proteins acquired during granulopoiesis and stored in multiple types of cytosolic granules with each granule type containing a unique cargo. Once released, granule proteins contribute to killing bacteria within the phagosome or the extracellular milieu, but are also capable of inflicting collateral tissue damage. Neutrophil-driven inflammation underlies many common diseases. Research over the last decade has documented neutrophil heterogeneity and functional versatility far beyond their antimicrobial function. Emerging evidence indicates that neutrophils utilize granule proteins to interact with innate and adaptive immune cells and orchestrate the inflammatory response. Granule proteins have been identified as important modulators of neutrophil trafficking, reverse transendothelial migration, phagocytosis, neutrophil life span, neutrophil extracellular trap formation, efferocytosis, cytokine activity, and autoimmunity. Hence, defining their roles within the inflammatory locus is critical for minimizing damage to the neighboring tissue and return to homeostasis. Here, we provide an overview of recent advances in the regulation of degranulation, granule protein functions, and signaling in modulating neutrophil-mediated immunity. We also discuss how targeting granule proteins and/or signaling could be harnessed for therapeutic benefits.
Collapse
Affiliation(s)
- Amira Othman
- Department of Pathology and Cell Biology, University of Montreal, QC, Canada.,Department of Biomedical Sciences, University of Montreal, QC, Canada.,Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| | - Meriem Sekheri
- Department of Biomedical Sciences, University of Montreal, QC, Canada.,Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| | - János G Filep
- Department of Pathology and Cell Biology, University of Montreal, QC, Canada.,Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| |
Collapse
|
140
|
Ring S, Inaba Y, Da M, Bopp T, Grabbe S, Enk A, Mahnke K. Regulatory T Cells Prevent Neutrophilic Infiltration of Skin during Contact Hypersensitivity Reactions by Strengthening the Endothelial Barrier. J Invest Dermatol 2021; 141:2006-2017. [PMID: 33675787 DOI: 10.1016/j.jid.2021.01.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
The healing phase of contact hypersensitivity reactions is critically dependent on regulatory T cells (Tregs), but even the early inflammatory phase, that is, 6-24 hours after induction of a contact hypersensitivity reaction, is susceptible to Treg-mediated suppression. To investigate the underlying mechanisms, we injected Tregs before the challenge and analyzed the skin-infiltrating cells as early as 6 hours later. Early on, we found mainly neutrophils in the challenged skin, but only a few T cells. This influx of neutrophils was blocked by the injection of Tregs, indicating that they were able to prevent the first wave of leukocytes, which are responsible for starting an immune reaction. As an underlying mechanism, we identified that Tregs can tighten endothelial junctions by inducing intracellular cAMP, leading to protein kinase A-RhoA‒dependent signaling. This eventually reorganizes endothelial junction proteins, such as Notch3, Nectin 2, Filamin B, and VE-cadherin, all of which contribute to the tightening of the endothelial barrier. In summary, Tregs prevent the leakage of proinflammatory cells from and into the tissue, which establishes a mechanism to downregulate immune reactions.
Collapse
Affiliation(s)
- Sabine Ring
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Yutaka Inaba
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Meihong Da
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Alexander Enk
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Karsten Mahnke
- Department of Dermatology, Heidelberg University Hospital, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany.
| |
Collapse
|
141
|
Vandendriessche S, Cambier S, Proost P, Marques PE. Complement Receptors and Their Role in Leukocyte Recruitment and Phagocytosis. Front Cell Dev Biol 2021; 9:624025. [PMID: 33644062 PMCID: PMC7905230 DOI: 10.3389/fcell.2021.624025] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/15/2021] [Indexed: 12/21/2022] Open
Abstract
The complement system is deeply embedded in our physiology and immunity. Complement activation generates a multitude of molecules that converge simultaneously on the opsonization of a target for phagocytosis and activation of the immune system via soluble anaphylatoxins. This response is used to control microorganisms and to remove dead cells, but also plays a major role in stimulating the adaptive immune response and the regeneration of injured tissues. Many of these effects inherently depend on complement receptors expressed on leukocytes and parenchymal cells, which, by recognizing complement-derived molecules, promote leukocyte recruitment, phagocytosis of microorganisms and clearance of immune complexes. Here, the plethora of information on the role of complement receptors will be reviewed, including an analysis of how this functionally and structurally diverse group of molecules acts jointly to exert the full extent of complement regulation of homeostasis.
Collapse
Affiliation(s)
- Sofie Vandendriessche
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Seppe Cambier
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Pedro E Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
142
|
Mechanisms of radiation-induced endothelium damage: Emerging models and technologies. Radiother Oncol 2021; 158:21-32. [PMID: 33581220 DOI: 10.1016/j.radonc.2021.02.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/24/2022]
Abstract
Radiation-induced endothelial/vascular injury is a major complicating factor in radiotherapy and a leading cause of morbidity and mortality in nuclear or radiological catastrophes. Exposure of tissue to ionizing radiation (IR) leads to the release of oxygen radicals and proteases that result in loss of endothelial barrier function and leukocyte dysfunction leading to tissue injury and organ damage. Microvascular endothelial cells are particularly sensitive to IR and radiation-induced alterations in endothelial cell function are thought to be a critical factor in organ damage through endothelial cell activation, enhanced leukocyte-endothelial cell interactions, increased barrier permeability and initiation of apoptotic pathways. These radiation-induced inflammatory responses are important in early and late radiation pathologies in various organs. A better understanding of mechanisms of radiation-induced endothelium dysfunction is therefore vital, as radiobiological response of endothelium is of major importance for medical management and therapeutic development for radiation injuries. In this review, we summarize the current knowledge of cellular and molecular mechanisms of radiation-induced endothelium damage and their impact on early and late radiation injury. Furthermore, we review established and emerging in vivo and in vitro models that have been developed to study the mechanisms of radiation-induced endothelium damage and to design, develop and rapidly screen therapeutics for treatment of radiation-induced vascular damage. Currently there are no specific therapeutics available to protect against radiation-induced loss of endothelial barrier function, leukocyte dysfunction and resulting organ damage. Developing therapeutics to prevent endothelium dysfunction and normal tissue damage during radiotherapy can serve as the urgently needed medical countermeasures.
Collapse
|
143
|
Boivin G, Ancey PB, Vuillefroy de Silly R, Kalambaden P, Contat C, Petit B, Ollivier J, Bourhis J, Meylan E, Vozenin MC. Anti-Ly6G binding and trafficking mediate positive neutrophil selection to unleash the anti-tumor efficacy of radiation therapy. Oncoimmunology 2021; 10:1876597. [PMID: 33628622 PMCID: PMC7889163 DOI: 10.1080/2162402x.2021.1876597] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 11/05/2022] Open
Abstract
The anti-Ly6G antibody is used to deplete Ly6Gpos neutrophils and study their role in diverse pathologies. However, depletion is never absolute, as Ly6Glow neutrophils resistant to depletion rapidly emerge. Studying the functionality of these residual neutrophils is necessary to interpret anti-Ly6G-based experimental designs. In vitro, we found anti-Ly6G binding induced Ly6G internalization, surface Ly6G paucity, and primed the oxidative burst of neutrophils upon TNF α co-stimulation. In vivo, we found neutrophils resistant to anti-Ly6G depletion exhibited anti-neutrophil-cytoplasmic-antibodies. In the pre-clinical KrasLox-STOP-Lox-G12D/WT; Trp53Flox/Flox mouse lung tumor model, abnormal neutrophil accumulation and aging was accompanied with an N2-like SiglecFpos polarization and ly6g downregulation. Consequently, SiglecFpos neutrophils exposed to anti-Ly6G reverted to Ly6Glow and were resistant to depletion. Noting that anti-Ly6G mediated neutrophil depletion alone had no anti-tumor effect, we found a long-lasting rate of tumor regression (50%) by combining anti-Ly6G with radiation-therapy, in this model reputed to be refractory to standard anticancer therapies. Mechanistically, anti-Ly6G regulated neutrophil aging while radiation-therapy enhanced the homing of anti-Ly6G-boundSiglecFneg neutrophils to tumors. This anti-tumor effect was recapitulated by G-CSF administration prior to RT and abrogated with an anti-TNFα antibody co-administration. In summary, we report that incomplete depletion of neutrophils using targeted antibodies can intrinsically promote their oxidative activity. This effect depends on antigen/antibody trafficking and can be harnessed locally using select delivery of radiation-therapy to impair tumor progression. This underutilized aspect of immune physiology may be adapted to expand the scope of neutrophil-related research.
Collapse
Affiliation(s)
- Gaël Boivin
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- School of Life Sciences Ecole Polytechnique Fédérale De, Lausanne, Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
- Radio-Oncology Service, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Pierre-Benoit Ancey
- School of Life Sciences Ecole Polytechnique Fédérale De, Lausanne, Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | | | - Pradeep Kalambaden
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Caroline Contat
- School of Life Sciences Ecole Polytechnique Fédérale De, Lausanne, Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Benoit Petit
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Radio-Oncology Service, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jonathan Ollivier
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Radio-Oncology Service, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jean Bourhis
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Radio-Oncology Service, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Etienne Meylan
- School of Life Sciences Ecole Polytechnique Fédérale De, Lausanne, Swiss Institute for Experimental Cancer Research, Lausanne, Switzerland
| | - Marie-Catherine Vozenin
- Radio-Oncology Laboratory, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Radio-Oncology Service, Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
144
|
|
145
|
Adams W, Espicha T, Estipona J. Getting Your Neutrophil: Neutrophil Transepithelial Migration in the Lung. Infect Immun 2021; 89:IAI.00659-20. [PMID: 33526562 DOI: 10.1128/iai.00659-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neutrophil transepithelial migration is a fundamental process that facilitates the rapid trafficking of neutrophils to inflammatory foci and occurs across a diverse range of tissues. For decades there has been widespread interest in understanding the mechanisms that drive this migratory process in response to different pathogens and organ systems. This has led to the successful integration of key findings on neutrophil transepithelial migration from the intestines, lungs, liver, genitourinary tract, and other tissues into a single, cohesive model. However, recent studies have identified organ specific differences in neutrophil transepithelial migration. These findings support a model where the tissue in concert with the pro-inflammatory stimuli dictate a unique collection of signals that drive neutrophil trafficking. This review focuses on the mechanisms that drive neutrophil transepithelial migration in response to microbial infection of a single organ, the lung. Herein we provide a detailed analysis of the adhesion molecules and chemoattractants that contribute to the recruitment of neutrophil into the airways. We also highlight important advances in experimental models for studying neutrophil transepithelial migration in the lung over the last decade.
Collapse
Affiliation(s)
- Walter Adams
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Taylor Espicha
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| | - Janine Estipona
- Department of Biological Sciences, San Jose State University, San Jose, CA 95192 USA
| |
Collapse
|
146
|
Heydarian M, Schweinlin M, Schwarz T, Rawal R, Walles H, Metzger M, Rudel T, Kozjak-Pavlovic V. Triple co-culture and perfusion bioreactor for studying the interaction between Neisseria gonorrhoeae and neutrophils: A novel 3D tissue model for bacterial infection and immunity. J Tissue Eng 2021; 12:2041731420988802. [PMID: 33796248 PMCID: PMC7970704 DOI: 10.1177/2041731420988802] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/31/2020] [Indexed: 01/13/2023] Open
Abstract
Gonorrhea, a sexually transmitted disease caused by the bacteria Neisseria gonorrhoeae, is characterized by a large number of neutrophils recruited to the site of infection. Therefore, proper modeling of the N. gonorrhoeae interaction with neutrophils is very important for investigating and understanding the mechanisms that gonococci use to evade the immune response. We have used a combination of a unique human 3D tissue model together with a dynamic culture system to study neutrophil transmigration to the site of N. gonorrhoeae infection. The triple co-culture model consisted of epithelial cells (T84 human colorectal carcinoma cells), human primary dermal fibroblasts, and human umbilical vein endothelial cells on a biological scaffold (SIS). After the infection of the tissue model with N. gonorrhoeae, we introduced primary human neutrophils to the endothelial side of the model using a perfusion-based bioreactor system. By this approach, we were able to demonstrate the activation and transmigration of neutrophils across the 3D tissue model and their recruitment to the site of infection. In summary, the triple co-culture model supplemented by neutrophils represents a promising tool for investigating N. gonorrhoeae and other bacterial infections and interactions with the innate immunity cells under conditions closely resembling the native tissue environment.
Collapse
Affiliation(s)
| | - Matthias Schweinlin
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Thomas Schwarz
- Translational Centre Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), Würzburg, Bayern, Germany
| | - Ravisha Rawal
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | - Heike Walles
- Research Center "Dynamic Systems: Systems Engineering" (CDS), Otto von-Guericke-University, Magdeburg, Sachsen-Anhalt, Germany
| | - Marco Metzger
- Translational Centre Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), Würzburg, Bayern, Germany
| | - Thomas Rudel
- Biocenter, Chair of Microbiology, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
147
|
CX3CR1 Depletion Promotes the Formation of Platelet-Neutrophil Complexes and Aggravates Acute Peritonitis. Shock 2021; 56:287-297. [PMID: 33481549 DOI: 10.1097/shk.0000000000001733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Peritonitis is a life-threatening condition on intensive care units. Inflammatory cytokines and their receptors drive inflammation, cause the formation of platelet-neutrophil complexes (PNCs) and therefore the migration of polymorphonuclear neutrophils (PMNs) into the inflamed tissue. CX3CL1 and its receptor CX3CR1 are expressed in various cells, and promote inflammation. The shedding of CX3CL1 is mediated by a disintegrin and metalloprotease (ADAM) 17. The role of the CX3CL1-CX3CR1 axis in acute peritonitis remains elusive. METHODS In zymosan-induced peritonitis, we determined the formation of PNCs in the blood and the expression of PNC-related molecules on PNCs. PMN migration into the peritoneal lavage was evaluated in wild-type (WT) and CX3CR1-/- animals by flow cytometry. CX3CL1, ADAM17, and the expression of various inflammatory cytokines were detected. Further, we determined the inflammation-associated activation of the intracellular transcription factor extracellular signal-regulated kinase 1/2 (ERK1/2) by Western blot. RESULTS The PMN accumulation in the peritoneal lavage and the PNC formation in the circulation were significantly raised in CX3CR1-/- compared with WT animals. The expression of PNC-related selectins on PNCs was significantly increased in the blood of CX3CR1-/- animals, as well as cytokine levels. Further, we observed an increased activation of ERK1/2 and elevated ADAM17 expression in CX3CR1-/- during acute inflammation. Selective ERK1/2 inhibition ameliorated inflammation-related increased ADAM17 expression. CONCLUSIONS A CX3CR1 deficiency raised the release of inflammatory cytokines and increased the PNC formation respectively PMN migration via an elevated ERK1/2 activation during acute peritonitis. Further, we observed a link between the ERK1/2 activation and an elevated ADAM17 expression on PNC-related platelets and PMNs during inflammation. Our data thus illustrate a crucial role of CX3CR1 on the formation of PNCs and regulating inflammation in acute peritonitis.
Collapse
|
148
|
Ulfig A, Leichert LI. The effects of neutrophil-generated hypochlorous acid and other hypohalous acids on host and pathogens. Cell Mol Life Sci 2021; 78:385-414. [PMID: 32661559 PMCID: PMC7873122 DOI: 10.1007/s00018-020-03591-y] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Neutrophils are predominant immune cells that protect the human body against infections by deploying sophisticated antimicrobial strategies including phagocytosis of bacteria and neutrophil extracellular trap (NET) formation. Here, we provide an overview of the mechanisms by which neutrophils kill exogenous pathogens before we focus on one particular weapon in their arsenal: the generation of the oxidizing hypohalous acids HOCl, HOBr and HOSCN during the so-called oxidative burst by the enzyme myeloperoxidase. We look at the effects of these hypohalous acids on biological systems in general and proteins in particular and turn our attention to bacterial strategies to survive HOCl stress. HOCl is a strong inducer of protein aggregation, which bacteria can counteract by chaperone-like holdases that bind unfolding proteins without the need for energy in the form of ATP. These chaperones are activated by HOCl through thiol oxidation (Hsp33) or N-chlorination of basic amino acid side-chains (RidA and CnoX) and contribute to bacterial survival during HOCl stress. However, neutrophil-generated hypohalous acids also affect the host system. Recent studies have shown that plasma proteins act not only as sinks for HOCl, but get actively transformed into modulators of the cellular immune response through N-chlorination. N-chlorinated serum albumin can prevent aggregation of proteins, stimulate immune cells, and act as a pro-survival factor for immune cells in the presence of cytotoxic antigens. Finally, we take a look at the emerging role of HOCl as a potential signaling molecule, particularly its role in neutrophil extracellular trap formation.
Collapse
Affiliation(s)
- Agnes Ulfig
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry-Microbial Biochemistry, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Lars I Leichert
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry-Microbial Biochemistry, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
149
|
Miralda I, Uriarte SM. Periodontal Pathogens' strategies disarm neutrophils to promote dysregulated inflammation. Mol Oral Microbiol 2020; 36:103-120. [PMID: 33128827 PMCID: PMC8048607 DOI: 10.1111/omi.12321] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/16/2020] [Accepted: 10/28/2020] [Indexed: 12/26/2022]
Abstract
Periodontitis is an irreversible, chronic inflammatory disease where inflammophilic pathogenic microbial communities accumulate in the gingival crevice. Neutrophils are a major component of the innate host response against bacterial challenge, and under homeostatic conditions, their microbicidal functions typically protect the host against periodontitis. However, a number of periodontal pathogens developed survival strategies to evade neutrophil microbicidal functions while promoting inflammation, which provides a source of nutrients for bacterial growth. Research on periodontal pathogens has largely focused on a few established species: Tannerella forsythia, Treponema denticola, Fusobacterium nucleatum, Aggregatibacter actinomycetemcomitans, and Porphyromonas gingivalis. However, advances in culture-independent techniques have facilitated the identification of new bacterial species in periodontal lesions, such as the two Gram-positive anaerobes, Filifactor alocis and Peptoanaerobacter stomatis, whose characterization of pathogenic potential has not been fully described. Additionally, there is not a full understanding of the pathogenic mechanisms used against neutrophils by organisms that are abundant in periodontal lesions. This presents a substantial barrier to the development of new approaches to prevent or ameliorate the disease. In this review, we first summarize the neutrophil functions affected by the established periodontal pathogens listed above, denoting unknown areas that still merit a closer look. Then, we review the literature on neutrophil functions and the emerging periodontal pathogens, F. alocis and P. stomatis, comparing the effects of the emerging microbes to that of established pathogens, and speculate on the contribution of these putative pathogens to the progression of periodontal disease.
Collapse
Affiliation(s)
- Irina Miralda
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| | - Silvia M Uriarte
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA
| |
Collapse
|
150
|
Chaconas G, Moriarty TJ, Skare J, Hyde JA. Live Imaging. Curr Issues Mol Biol 2020; 42:385-408. [PMID: 33310914 PMCID: PMC7946808 DOI: 10.21775/cimb.042.385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Being able to vizualize a pathogen at a site of interaction with a host is an aesthetically appealing idea and the resulting images can be both informative as well as enjoyable to view. Moreover, the approaches used to derive these images can be powerful in terms of offering data unobtainable by other methods. In this article, we review three primary modalities for live imaging Borrelia spirochetes: whole animal imaging, intravital microscopy and live cell imaging. Each method has strengths and weaknesses, which we review, as well as specific purposes for which they are optimally utilized. Live imaging borriliae is a relatively recent development and there was a need of a review to cover the area. Here, in addition to the methods themselves, we also review areas of spirochete biology that have been significantly impacted by live imaging and present a collection of images associated with the forward motion in the field driven by imaging studies.
Collapse
Affiliation(s)
- George Chaconas
- Department of Biochemistry and Molecular Biology and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, T2N 4N1, Canada
| | - Tara J. Moriarty
- Faculties of Dentistry and Medicine (Laboratory Medicine and Pathobiology), University of Toronto, Toronto, Ontario, M5G 1G6, Canada
| | - Jon Skare
- Department of Microbial Pathogenesis and Immunology, Texas A & M University Health Science Center, Bryan, Texas, 77807, USA
| | - Jenny A. Hyde
- Department of Microbial Pathogenesis and Immunology, Texas A & M University Health Science Center, Bryan, Texas, 77807, USA
| |
Collapse
|