101
|
Kapoor D, Shukla D. Neutrophil Extracellular Traps and Their Possible Implications in Ocular Herpes Infection. Pathogens 2023; 12:209. [PMID: 36839481 PMCID: PMC9958879 DOI: 10.3390/pathogens12020209] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/01/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are net-like structures released from neutrophils. NETs predominantly contain cell-free deoxyribonucleic acid (DNA) decorated with histones and neutrophil granule proteins. Numerous extrinsic and intrinsic stimuli can induce the formation of NETs such as pathogens, cytokines, immune complexes, microcrystals, antibodies, and other physiological stimuli. The mechanism of NETosis induction can either be ROS-dependent or independent based on the catalase producing activity of the pathogen. NADPH is the source of ROS production, which in turn depends on the upregulation of Ca2+ production in the cytoplasm. ROS-independent induction of NETosis is regulated through toll-like receptors (TLRs). Besides capturing and eliminating pathogens, NETs also aggravate the inflammatory response and thus act as a double-edged sword. Currently, there are growing reports of NETosis induction during bacterial and fungal ocular infections leading to different pathologies, but there is no direct report suggesting its role during herpes simplex virus (HSV) infection. There are innumerable independent reports showing that the major effectors of NETosis are also directly affected by HSV infection, and thus, there is a strong possibility that HSV interacts with these facilitators that can either result in virally mediated modulation of NETosis or NETosis-mediated suppression of ocular HSV infection. This review focuses on the mechanism of NETs formation during different ocular pathologies, with its prime focus on highlighting their potential implications during HSV ocular infections and acting as prospective targets for the treatment of ocular diseases.
Collapse
Affiliation(s)
- Divya Kapoor
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1905 W. Taylor St., Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
| | - Deepak Shukla
- Department of Ophthalmology and Visual Sciences, College of Medicine, University of Illinois at Chicago, 1905 W. Taylor St., Chicago, IL 60612, USA
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, 835 S. Wolcott, Chicago, IL 60612, USA
| |
Collapse
|
102
|
Yan S, Li M, Liu B, Ma Z, Yang Q. Neutrophil extracellular traps and pulmonary fibrosis: an update. J Inflamm (Lond) 2023; 20:2. [PMID: 36658568 PMCID: PMC9851107 DOI: 10.1186/s12950-023-00329-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Pulmonary fibrosis (PF) is a serious and often fatal illness that occurs in various clinical settings and represents a significant unmet medical need. Increasing evidence indicates that neutrophil extracellular traps (NETs) contribute significantly to the progression of PF. Therefore, understanding the pathways by which NETs contribute to the disease is crucial for developing effective treatments. This review focuses on the formation of NETs and the common mechanisms of NETs in PF.
Collapse
Affiliation(s)
- Suyan Yan
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Meiqi Li
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Baocheng Liu
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Zhenzhen Ma
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| | - Qingrui Yang
- grid.460018.b0000 0004 1769 9639Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Huaiyin District, Jinan, 250021 Shandong China
| |
Collapse
|
103
|
Hudock KM, Collins MS, Imbrogno MA, Kramer EL, Brewington JJ, Ziady A, Zhang N, Snowball J, Xu Y, Carey BC, Horio Y, O’Grady SM, Kopras EJ, Meeker J, Morgan H, Ostmann AJ, Skala E, Siefert ME, Na CL, Davidson CR, Gollomp K, Mangalmurti N, Trapnell BC, Clancy JP. Alpha-1 antitrypsin limits neutrophil extracellular trap disruption of airway epithelial barrier function. Front Immunol 2023; 13:1023553. [PMID: 36703990 PMCID: PMC9872031 DOI: 10.3389/fimmu.2022.1023553] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/30/2022] [Indexed: 01/12/2023] Open
Abstract
Neutrophil extracellular traps contribute to lung injury in cystic fibrosis and asthma, but the mechanisms are poorly understood. We sought to understand the impact of human NETs on barrier function in primary human bronchial epithelial and a human airway epithelial cell line. We demonstrate that NETs disrupt airway epithelial barrier function by decreasing transepithelial electrical resistance and increasing paracellular flux, partially by NET-induced airway cell apoptosis. NETs selectively impact the expression of tight junction genes claudins 4, 8 and 11. Bronchial epithelia exposed to NETs demonstrate visible gaps in E-cadherin staining, a decrease in full-length E-cadherin protein and the appearance of cleaved E-cadherin peptides. Pretreatment of NETs with alpha-1 antitrypsin (A1AT) inhibits NET serine protease activity, limits E-cadherin cleavage, decreases bronchial cell apoptosis and preserves epithelial integrity. In conclusion, NETs disrupt human airway epithelial barrier function through bronchial cell death and degradation of E-cadherin, which are limited by exogenous A1AT.
Collapse
Affiliation(s)
- K. M. Hudock
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States,*Correspondence: K. M. Hudock,
| | - M. S. Collins
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - M. A. Imbrogno
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - E. L. Kramer
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Division of Pediatric Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - J. J. Brewington
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Division of Pediatric Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - A. Ziady
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - N. Zhang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - J. Snowball
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Y. Xu
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Divisions of Biomedical Informatics, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - B. C. Carey
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Y. Horio
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Department of Respiratory Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto-shi, Kumamoto, Japan
| | - S. M. O’Grady
- Departments of Animal Science, University of Minnesota, St. Paul, MN, United States,Department of Integrative Biology and Physiology, University of Minnesota, St. Paul, MN, United States
| | - E. J. Kopras
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - J. Meeker
- Division of Pediatric Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - H. Morgan
- Division of Pediatric Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - A. J. Ostmann
- Division of Pediatric Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - E. Skala
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - M. E. Siefert
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - C. L. Na
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - C. R. Davidson
- Division of Pediatric Pulmonary Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - K. Gollomp
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States,Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - N. Mangalmurti
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States,Pennsylvania Lung Biology Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - B. C. Trapnell
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States,Translational Pulmonary Science Center, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - J. P. Clancy
- Cystic Fibrosis Foundation, Bethesda, MD, United States
| |
Collapse
|
104
|
Salinas C, Barriga K, Albornoz A, Alarcon P, Quiroga J, Uberti B, Sarmiento J, Henriquez C, Ehrenfeld P, Burgos RA, Moran G. Tamoxifen triggers the in vitro release of neutrophil extracellular traps in healthy horses. Front Vet Sci 2023; 9:1025249. [PMID: 36686170 PMCID: PMC9853556 DOI: 10.3389/fvets.2022.1025249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/08/2022] [Indexed: 01/07/2023] Open
Abstract
Neutrophils display an array of biological functions including the formation of neutrophil extracellular traps (NETs), web-like structures specialized in trapping, neutralizing, killing and preventing microbial dissemination within the host. However, NETs contribute to a number of inflammatory pathologies, including severe equine asthma. Tamoxifen (TX) is a selective estrogen receptor modulator which belongs to the triphenylethyllenes group of molecules, and which is used as a treatment in all stages of estrogen-positive human breast cancer. Our previous results suggest that tamoxifen can modulate neutrophil functionality and promote resolution of inflammation; this would partly explain the clinical beneficial effect of this drug in horses with airway inflammation. Enhanced NETs production has been reported with tamoxifen use in humans, but minimal data exists regarding the drug's effect on NETs in horses. The aim of this study is to assess the in vitro effect of TX on NETs formation from peripheral blood of healthy horses. Five clinically healthy mixed-breed adult horses were enrolled in the study. For this, cellular free DNA quantification, immunofluorescence for the visualization of NETs, assessment of different types of NETs, and detection of mitochondrial superoxide. TX induced NETs formation at a concentration of 10 uM. Our results show that only two types of NETs were induced by TX: 95% spread NETs (sprNETs) and 5% aggregated NETs (aggNETs). Furthermore, induction of these NETs could be influenced by mitochondrial ROS. Future research should involve an In vivo study of horses with severe asthma and TX treatment, to evaluate BALF neutrophil NET formation. In conclusion, this in vitro study suggests that the resolution of inflammation by TX in horses with airway inflammation is due to inhibition of other neutrophilic functions but not to NET formation.
Collapse
Affiliation(s)
- Constanza Salinas
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Kassandra Barriga
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Alejandro Albornoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Pablo Alarcon
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - John Quiroga
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Benjamín Uberti
- Instituto de Ciencias Clínicas Veterinarias, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - José Sarmiento
- Instituto de Fisiología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Claudio Henriquez
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Instituto de Anatomía, Histología y Patología, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael A. Burgos
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Gabriel Moran
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile,*Correspondence: Gabriel Moran ✉
| |
Collapse
|
105
|
Gong HH, Worley MJ, Carver KA, Goldstein DR, Deng JC. Neutrophils drive pulmonary vascular leakage in MHV-1 infection of susceptible A/J mice. Front Immunol 2023; 13:1089064. [PMID: 36685578 PMCID: PMC9853883 DOI: 10.3389/fimmu.2022.1089064] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023] Open
Abstract
Background Lung inflammation, neutrophil infiltration, and pulmonary vascular leakage are pathological hallmarks of acute respiratory distress syndrome (ARDS) which can lethally complicate respiratory viral infections. Despite similar comorbidities, however, infections in some patients may be asymptomatic while others develop ARDS as seen with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections for example. Methods In this study, we infected resistant C57BL/6 and susceptible A/J strains of mice with pulmonary administration of murine hepatitis virus strain 1 (MHV-1) to determine mechanisms underlying susceptibility to pulmonary vascular leakage in a respiratory coronavirus infection model. Results A/J animals displayed increased lung injury parameters, pulmonary neutrophil influx, and deficient recruitment of other leukocytes early in the infection. Moreover, under basal conditions, A/J neutrophils overexpressed primary granule protein genes for myeloperoxidase and multiple serine proteases. During infection, myeloperoxidase and elastase protein were released in the bronchoalveolar spaces at higher concentrations compared to C57BL/6 mice. In contrast, genes from other granule types were not differentially expressed between these 2 strains. We found that depletion of neutrophils led to mitigation of lung injury in infected A/J mice while having no effect in the C57BL/6 mice, demonstrating that an altered neutrophil phenotype and recruitment profile is a major driver of lung immunopathology in susceptible mice. Conclusions These results suggest that host susceptibility to pulmonary coronaviral infections may be governed in part by underlying differences in neutrophil phenotypes, which can vary between mice strains, through mechanisms involving primary granule proteins as mediators of neutrophil-driven lung injury.
Collapse
Affiliation(s)
- Henry H. Gong
- University of Michigan, Ann Arbor, MI, United States
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Matthew J. Worley
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Kyle A. Carver
- Research Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Daniel R. Goldstein
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Jane C. Deng
- University of Michigan, Ann Arbor, MI, United States
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
- Medicine Service, Veterans Affairs (VA) Ann Arbor Healthcare System, Department of Veterans Affairs Health System, Ann Arbor, MI, United States
| |
Collapse
|
106
|
Yili S, Xinyi D, Kerui F, Kun C, Yang Y, Zhang L, Hu K. Activation of GPR81 aggravated intestinal ischemia/reperfusion injury-induced acute lung injury via HMGB1-mediated neutrophil extracellular traps formation. Int J Immunopathol Pharmacol 2023; 37:3946320231193832. [PMID: 37698122 PMCID: PMC10498694 DOI: 10.1177/03946320231193832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 07/25/2023] [Indexed: 09/13/2023] Open
Abstract
INTRODUCTION Intestinal ischemia/reperfusion (II/R) injury is a life-threatening situation accompanied by severe organ injury, especially acute lung injury (ALI). A great body of evidence indicates that II/R injury is usually associated with hyperlactatemia. G-protein-coupled receptor 81 (GPR81), a receptor of lactate, has been recognized as a regulatory factor in inflammation, but whether it was involved in II/R injury-induced ALI is still unknown. METHODS To establish the II/R injury model, the superior mesenteric artery of the mice was occluded gently by a microvascular clamp for 45 min to elicit intestinal ischemia and then a 90-min reperfusion was performed. Broncho-alveolar lavage fluid (BALF) and lung tissues were obtained to evaluate the lung injury after II/R. The pulmonary histopathological alteration was evaluated by H&E staining. The concentration of proteins, the number of infiltrated cells, and the level of IL-6 were measured in BALF. The formation of neutrophil extracellular traps (NETs) was evaluated by the level of double-stranded DNA (dsDNA) and myeloperoxidase- double-stranded DNA (MPO-dsDNA) complex in BALF, and the content of citrullinated histone H3 (Cit-H3) in lung tissue. The level of HMGB1 in the BALF and plasma was measured by enzyme linked immunosorbent assay (ELISA). RESULTS Administration of the GPR81 agonist 3,5-dihydroxybenzoic acid (DHBA) aggravated II/R injury-induced lung histological abnormalities, upregulated the concentration of proteins, the number of infiltrated cells, and the level of IL-6 in BALF. In addition, DHBA treatment increased the level of dsDNA and MPO-dsDNA complex in BALF, and promoted the elevation of Cit-H3 in lung tissue and the release of HMGB1 in BALF and plasma. CONCLUSION After induction of ALI by II/R, the administration of DHBA aggravated ALI through NETs formation in the lung.
Collapse
Affiliation(s)
- Sun Yili
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Dai Xinyi
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Fan Kerui
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Chen Kun
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Yongqiang Yang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Li Zhang
- Department of Pathophysiology, Chongqing Medical University, Chongqing, China
| | - Kai Hu
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, China
| |
Collapse
|
107
|
Thomas PG, Shubina M, Balachandran S. ZBP1/DAI-Dependent Cell Death Pathways in Influenza A Virus Immunity and Pathogenesis. Curr Top Microbiol Immunol 2023; 442:41-63. [PMID: 31970498 DOI: 10.1007/82_2019_190] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Influenza A viruses (IAV) are members of the Orthomyxoviridae family of negative-sense RNA viruses. The greatest diversity of IAV strains is found in aquatic birds, but a subset of strains infects other avian as well as mammalian species, including humans. In aquatic birds, infection is largely restricted to the gastrointestinal tract and spread is through feces, while in humans and other mammals, respiratory epithelial cells are the primary sites supporting productive replication and transmission. IAV triggers the death of most cell types in which it replicates, both in culture and in vivo. When well controlled, such cell death is considered an effective host defense mechanism that eliminates infected cells and limits virus spread. Unchecked or inopportune cell death also results in immunopathology. In this chapter, we discuss the impact of cell death in restricting virus spread, supporting the adaptive immune response and driving pathogenesis in the mammalian respiratory tract. Recent studies have begun to shed light on the signaling pathways underlying IAV-activated cell death. These pathways, initiated by the pathogen sensor protein ZBP1 (also called DAI and DLM1), cause infected cells to undergo apoptosis, necroptosis, and pyroptosis. We outline mechanisms of ZBP1-mediated cell death signaling following IAV infection.
Collapse
Affiliation(s)
- Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, MS 351, 262 Danny Thomas Place, 38105, Memphis, TN, USA.
| | - Maria Shubina
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA
| | - Siddharth Balachandran
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Room 224 Reimann Building, 333 Cottman Ave., 19111, Philadelphia, PA, USA.
| |
Collapse
|
108
|
Xie J, Zhu CL, Wan XJ, Zhao ZZ, Meng Y, Li P, Guo Y, Liu Q, Bian JJ, Deng XM, Wang JF. GSDMD-mediated NETosis promotes the development of acute respiratory distress syndrome. Eur J Immunol 2023; 53:e2250011. [PMID: 36250416 DOI: 10.1002/eji.202250011] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 02/02/2023]
Abstract
Gasdermin D (GSDMD) is a classical molecule involved in pyroptosis. It has been reported to be cleaved into N-terminal fragments to form pores in the neutrophil membrane and promote the release of neutrophil extracellular traps (NETs). However, it remains unclear if GSDMD is involved in neutrophil regulation and NET release during ARDS. The role of neutrophil GSDMD in the development of ARDS was investigated in a murine model of ARDS induced by lipopolysaccharide (LPS) using the neutrophil specific GSDMD-deficient mice. The neutrophil GSDMD cleavage and its relationship with NETosis were also explored in ARDS patients. The cleavage of GSDMD in neutrophils from ARDS patients and mice was upregulated. Inhibition of GSDMD by genetic knockout or inhibitors resulted in reduced production of NET both in vivo and in vitro, and attenuation of LPS-induced lung injury. Moreover, in vitro experiments showed that the inhibition of GSDMD attenuated endothelial injury co-cultured with neutrophils from ARDS patients, while extrinsic NETs reversed the protective effect of GSDMD inhibition. Collectively, our data suggest that the neutrophil GSDMD cleavage is crucial in NET release during ARDS. The NET release maintained by cleaved GSDMD in neutrophils may be a key event in the development of ARDS.
Collapse
Affiliation(s)
- Jian Xie
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Cheng-Long Zhu
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Xiao-Jian Wan
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Zhen-Zhen Zhao
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Yan Meng
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Peng Li
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Yu Guo
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Qiang Liu
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Jin-Jun Bian
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Xiao-Ming Deng
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| | - Jia-Feng Wang
- Faculty of Anesthesiology, Changhai Hospital, the Naval Medical University, Shanghai, P. R. China
| |
Collapse
|
109
|
Luo H, Guo H, Zhou Y, Fang R, Zhang W, Mei Z. Neutrophil Extracellular Traps in Cerebral Ischemia/Reperfusion Injury: Friend and Foe. Curr Neuropharmacol 2023; 21:2079-2096. [PMID: 36892020 PMCID: PMC10556361 DOI: 10.2174/1570159x21666230308090351] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/19/2022] [Accepted: 12/26/2022] [Indexed: 03/10/2023] Open
Abstract
Cerebral ischemic injury, one of the leading causes of morbidity and mortality worldwide, triggers various central nervous system (CNS) diseases, including acute ischemic stroke (AIS) and chronic ischemia-induced Alzheimer's disease (AD). Currently, targeted therapies are urgently needed to address neurological disorders caused by cerebral ischemia/reperfusion injury (CI/RI), and the emergence of neutrophil extracellular traps (NETs) may be able to relieve the pressure. Neutrophils are precursors to brain injury following ischemic stroke and exert complicated functions. NETs extracellularly release reticular complexes of neutrophils, i.e., double-stranded DNA (dsDNA), histones, and granulins. Paradoxically, NETs play a dual role, friend and foe, under different conditions, for example, physiological circumstances, infection, neurodegeneration, and ischemia/reperfusion. Increasing evidence indicates that NETs exert anti-inflammatory effects by degrading cytokines and chemokines through protease at a relatively stable and moderate level under physiological conditions, while excessive amounts of NETs release (NETosis) irritated by CI/RI exacerbate the inflammatory response and aggravate thrombosis, disrupt the blood-brain barrier (BBB), and initiates sequential neuron injury and tissue damage. This review provides a comprehensive overview of the machinery of NETs formation and the role of an abnormal cascade of NETs in CI/RI, as well as other ischemia-induced neurological diseases. Herein, we highlight the potential of NETs as a therapeutic target against ischemic stroke that may inspire translational research and innovative clinical approaches.
Collapse
Affiliation(s)
- Haoyue Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Hanjing Guo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Yue Zhou
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Rui Fang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| |
Collapse
|
110
|
Lundstrom K, Hromić-Jahjefendić A, Bilajac E, Aljabali AAA, Baralić K, Sabri NA, Shehata EM, Raslan M, Ferreira ACBH, Orlandi L, Serrano-Aroca Á, Tambuwala MM, Uversky VN, Azevedo V, Alzahrani KJ, Alsharif KF, Halawani IF, Alzahrani FM, Redwan EM, Barh D. COVID-19 signalome: Pathways for SARS-CoV-2 infection and impact on COVID-19 associated comorbidity. Cell Signal 2023; 101:110495. [PMID: 36252792 PMCID: PMC9568271 DOI: 10.1016/j.cellsig.2022.110495] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 01/08/2023]
Abstract
The COVID-19 pandemic has been the focus of research the past two years. The major breakthrough was made by discovering pathways related to SARS-CoV-2 infection through cellular interaction by angiotensin-converting enzyme (ACE2) and cytokine storm. The presence of ACE2 in lungs, intestines, cardiovascular tissues, brain, kidneys, liver, and eyes shows that SARS-CoV-2 may have targeted these organs to further activate intracellular signalling pathways that lead to cytokine release syndrome. It has also been reported that SARS-CoV-2 can hijack coatomer protein-I (COPI) for S protein retrograde trafficking to the endoplasmic reticulum-Golgi intermediate compartment (ERGIC), which, in turn, acts as the assembly site for viral progeny. In infected cells, the newly synthesized S protein in endoplasmic reticulum (ER) is transported first to the Golgi body, and then from the Golgi body to the ERGIC compartment resulting in the formation of specific a motif at the C-terminal end. This review summarizes major events of SARS-CoV-2 infection route, immune response following host-cell infection as an important factor for disease outcome, as well as comorbidity issues of various tissues and organs arising due to COVID-19. Investigations on alterations of host-cell machinery and viral interactions with multiple intracellular signaling pathways could represent a major factor in more effective disease management.
Collapse
Affiliation(s)
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina.
| | - Esma Bilajac
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka Cesta 15, 71000 Sarajevo, Bosnia and Herzegovina
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid 21163, Jordan.
| | - Katarina Baralić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Nagwa A Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11865, Egypt.
| | - Eslam M Shehata
- Drug Research Center, Clinical Research and Bioanalysis Department, Cairo 11865, Egypt.
| | - Mohamed Raslan
- Drug Research Center, Clinical Research and Bioanalysis Department, Cairo 11865, Egypt.
| | - Ana Cláudia B H Ferreira
- Campinas State University, Campinas, São Paulo, Brazil; University Center of Lavras (UNILAVRAS), Lavras, Minas Gerais, Brazil.
| | - Lidiane Orlandi
- University Center of Lavras (UNILAVRAS), Lavras, Minas Gerais, Brazil.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Laboratory, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain.
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Vasco Azevedo
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Khalid J Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Khalaf F Alsharif
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Ibrahim F Halawani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Fuad M Alzahrani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, P.O. Box 80203, Jeddah, Saudi Arabia.
| | - Debmalya Barh
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil; Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur 721172, India.
| |
Collapse
|
111
|
Xu Y, Liu X, Zhang Z. STV-Na attenuates lipopolysaccharide-induced lung injury in mice via the TLR4/NF-kB pathway. Immun Inflamm Dis 2023; 11:e770. [PMID: 36705406 PMCID: PMC9846117 DOI: 10.1002/iid3.770] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI) is a potentially fatal disorder that is largely caused by inflammation. Sodium isostevanol (STV-Na) is a terpenoid produced from stevioside, which possesses anti-inflammatory and antioxidative stress characteristics. nevertheless, it is still unclear how STV-Na affects ALI. Therefore, we investigated the possible STV-Na therapeutic impacts on lipopolysaccharide (LPS)-induced (ALI). METHODS We employed hematoxylin-eosin staining to observe the impact of STV-Na on lung histopathological alterations and used kits to detect the oxidative stress status of lung tissues, such as superoxide dismutase, malondialdehyde, and glutathione. The reactive oxygen species and myeloperoxidase expression in the tissues of lung was assessed by immunofluorescence and immunohistochemistry. Additionally, we detected the impact of STV-Na on inflammatory cell infiltration in lung tissue using Wright-Giemsa staining solution and immunohistochemistry, which was found to reduce inflammation in lung tissue by enzyme-linked immunosorbent assay. Finally, using WB, we examined the impact of STV-Na on the TLR4/NF-kB pathway. RESULTS We observed that STV-Na attenuated lung histopathological alterations in LPS-induced lung damage in mice, reduced infiltration of inflammatory cell and oxidative stress in the tissue of lung, and via the TLR4/NF-kB pathway, there is a reduction in the inflammatory responses in mouse lung tissue. CONCLUSIONS These outcomes indicate that the response of inflammatory cells to LPS-induced ALI in mice was attenuated by STV-Na.
Collapse
Affiliation(s)
- Yanhong Xu
- Department of RespiratoryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Xiaoming Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Xinjiang Medical UniversityXinjiang Medical UniversityXinjiangUrumqiChina
| | - Zhihui Zhang
- Department of Plastic Surgery, The First Affiliated Hospital of Xinjiang Medical UniversityXinjiang Medical UniversityXinjiangUrumqiChina
| |
Collapse
|
112
|
Abstract
AIM Acute lung injury (ALI) is a common complication of severe acute pancreatitis (SAP) with a high mortality. Early prediction of patients at risk in initial stage can improve the long-term survival. METHODS A total of 91 patients with SAP out of 1647 acute pancreatitis patients from January 2015 to December 2020 were considered. A predictive model for SAP-associated ALI was constructed based on the valuable risk factors identified from routine clinical characteristics and plasma biomarkers. The value of the model was evaluated and compared with Lung Injury Prediction Score (LIPS). A nomogram was built to visualize the model. RESULTS Diabetes, oxygen supplementation, neutrophil count and D-dimer were found to be associated with ALI in SAP. The predictive model based on these factors had an area under the receiver operating characteristic curve [AUC: 0.88, 95% confidence interval (CI): 0.81-0.95], which was superior to LIPS (AUC: 0.71, 95% CI: 0.60-0.83), also with the higher sensitivity (65%) and specificity (96%) than LIPS (62%, 74%, respectively). Decision curve analysis of the model showed a higher net benefit than LIPS. Visualization by a nomogram facilitated the application of the model. CONCLUSION Diabetes, oxygen supplementation, neutrophil count and D-dimer were risk factors for SAP-associated ALI. The combination of these routine clinical data and the model visualization by a nomogram provided a simple and effective way in predicting ALI in the early phase of SAP.
Collapse
|
113
|
COVID-19 Pathology Sheds Further Light on Balance between Neutrophil Proteases and Their Inhibitors. Biomolecules 2022; 13:biom13010082. [PMID: 36671467 PMCID: PMC9855895 DOI: 10.3390/biom13010082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Excessive neutrophil influx and activation in lungs during infections, such as manifest during the ongoing SARS CoV-2 pandemic, have brought neutrophil extracellular traps (NETs) and the concomitant release of granule contents that damage surrounding tissues into sharp focus. Neutrophil proteases, which are known to participate in NET release, also enable the binding of the viral spike protein to cellular receptors and assist in the spread of infection. Blood and tissue fluids normally also contain liver-derived protease inhibitors that balance the activity of proteases. Interestingly, neutrophils themselves also express the protease inhibitor alpha-1-antitrypsin (AAT), the product of the SERPINA-1 gene, and store it in neutrophil cytoplasmic granules. The absence of AAT or mutations in the SERPINA-1 gene promotes lung remodeling and fibrosis in diseases such as chronic obstructive pulmonary disease (COPD), and acute respiratory distress syndrome (ARDS) and increases the risk of allergic responses. Recent observations point to the fact that reduced activity of AAT presents a major susceptibility factor for severe COVID-19. Here, we focus attention on the mechanism of neutrophil elastase (NE) in NET release and its inhibition by AAT as an additional factor that may determine the severity of COVID-19.
Collapse
|
114
|
Cardelli M, Pierpaoli E, Marchegiani F, Marcheselli F, Piacenza F, Giacconi R, Recchioni R, Casoli T, Stripoli P, Provinciali M, Matacchione G, Giuliani A, Ramini D, Sabbatinelli J, Bonafè M, Di Rosa M, Cherubini A, Di Pentima C, Spannella F, Antonicelli R, Bonfigli AR, Olivieri F, Lattanzio F. Biomarkers of cell damage, neutrophil and macrophage activation associated with in-hospital mortality in geriatric COVID-19 patients. Immun Ageing 2022; 19:65. [PMID: 36522763 PMCID: PMC9751505 DOI: 10.1186/s12979-022-00315-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 10/10/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND The risk for symptomatic COVID-19 requiring hospitalization is higher in the older population. The course of the disease in hospitalised older patients may show significant variation, from mild to severe illness, ultimately leading to death in the most critical cases. The analysis of circulating biomolecules involved in mechanisms of inflammation, cell damage and innate immunity could lead to identify new biomarkers of COVID-19 severity, aimed to improve the clinical management of subjects at higher risk of severe outcomes. In a cohort of COVID-19 geriatric patients (n= 156) who required hospitalization we analysed, on-admission, a series of circulating biomarkers related to neutrophil activation (neutrophil elastase, LL-37), macrophage activation (sCD163) and cell damage (nuclear cfDNA, mithocondrial cfDNA and nuclear cfDNA integrity). The above reported biomarkers were tested for their association with in-hospital mortality and with clinical, inflammatory and routine hematological parameters. Aim of the study was to unravel prognostic parameters for risk stratification of COVID-19 patients. RESULTS Lower n-cfDNA integrity, higher neutrophil elastase and higher sCD163 levels were significantly associated with an increased risk of in-hospital decease. Median (IQR) values observed in discharged vs. deceased patients were: 0.50 (0.30-0.72) vs. 0.33 (0.22-0.62) for n-cfDNA integrity; 94.0 (47.7-154.0) ng/ml vs. 115.7 (84.2-212.7) ng/ml for neutrophil elastase; 614.0 (370.0-821.0) ng/ml vs. 787.0 (560.0-1304.0) ng/ml for sCD163. The analysis of survival curves in patients stratified for tertiles of each biomarker showed that patients with n-cfDNA integrity < 0.32 or sCD163 in the range 492-811 ng/ml had higher risk of in-hospital decease than, respectively, patients with higher n-cfDNA integrity or lower sCD163. These associations were further confirmed in multivariate models adjusted for age, sex and outcome-related clinical variables. In these models also high levels of neutrophil elastase (>150 ng/ml) appeared to be independent predictor of in-hospital death. An additional analysis of neutrophil elastase in patients stratified for n-cfDNA integrity levels was conducted to better describe the association of the studied parameters with the outcome. CONCLUSIONS On the whole, biomarkers of cell-free DNA integrity, neutrophil and macrophage activation might provide a valuable contribution to identify geriatric patients with high risk of COVID-19 in-hospital mortality.
Collapse
Affiliation(s)
- M. Cardelli
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - E. Pierpaoli
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - F. Marchegiani
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - F. Marcheselli
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - F. Piacenza
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - R. Giacconi
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - R. Recchioni
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - T. Casoli
- Center for Neurobiology of Aging, Scientific Technological Area, IRCCS INRCA, Via Birarelli 8, 60121 Ancona, Italy
| | - P. Stripoli
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - M. Provinciali
- Advanced Technology Center for Aging Research, Scientific Technological Area, IRCCS INRCA, Ancona, Italy
| | - G. Matacchione
- grid.7010.60000 0001 1017 3210Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - A. Giuliani
- grid.7010.60000 0001 1017 3210Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - D. Ramini
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - J. Sabbatinelli
- grid.411490.90000 0004 1759 6306SOD Medicina di Laboratorio, Azienda Ospedaliero Universitaria Ospedali Riuniti, Ancona, Italy
| | - M. Bonafè
- grid.6292.f0000 0004 1757 1758Department of Experimental, Diagnostic, and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - M. Di Rosa
- Unit of Geriatric Pharmacoepidemiology and Biostatistics, IRCCS INRCA, Cosenza, Italy
| | - A. Cherubini
- Geriatria, Accettazione geriatrica e Centro di Ricerca per l’invecchiamento, IRCCS INRCA, Ancona, Italy
| | - C. Di Pentima
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy
| | - F. Spannella
- Internal Medicine and Geriatrics, IRCCS INRCA, Via della Montagnola 81, 60127 Ancona, Italy
| | | | - A. R. Bonfigli
- Scientific Direction and Geriatric Unit, IRCCS INRCA, Ancona, Italy
| | - F. Olivieri
- grid.7010.60000 0001 1017 3210Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Via Tronto 10/a, 60126 Ancona, Italy
| | - F. Lattanzio
- Scientific Direction and Geriatric Unit, IRCCS INRCA, Ancona, Italy
| |
Collapse
|
115
|
de Jesus Gonzalez-Contreras F, Zarate X. Neutrophil extracellular traps: Modulation mechanisms by pathogens. Cell Immunol 2022; 382:104640. [PMID: 36413806 DOI: 10.1016/j.cellimm.2022.104640] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/11/2022]
Abstract
Neutrophils, as innate effector cells, play an essential role in the containment and elimination of pathogens. Among the main neutrophil mechanisms use for these processes is the release of neutrophil extracellular traps (NETs), which consist of decondensed DNA decorated with various cytoplasmic proteins. NETs' principal role is the trapping and elimination of infectious agents; therefore, the formation of NETs is regulated by bacteria, fungi, parasites, and viruses through different mechanisms: the presence of virulence factors (adhered or secreted), microbial load, size of the microorganism, and even due to other immune cells activation (mainly platelets). This review summarizes the significant aspects that contribute to NETs modulation by pathogens and their components, and the effect NETs have on these pathogens as a cellular defense mechanism.
Collapse
Affiliation(s)
| | - Xristo Zarate
- Facultad de Ciencias Quimicas, Universidad Autonoma de Nuevo Leon, Av. Universidad s/n, San Nicolas de los Garza 66455, NL, Mexico
| |
Collapse
|
116
|
Enomoto N. Pathological Roles of Pulmonary Cells in Acute Lung Injury: Lessons from Clinical Practice. Int J Mol Sci 2022; 23:ijms232315027. [PMID: 36499351 PMCID: PMC9736972 DOI: 10.3390/ijms232315027] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/05/2022] Open
Abstract
Interstitial lung diseases (ILD) are relatively rare and sometimes become life threatening. In particular, rapidly progressive ILD, which frequently presents as acute lung injury (ALI) on lung histopathology, shows poor prognosis if proper and immediate treatments are not initiated. These devastating conditions include acute exacerbation of idiopathic pulmonary fibrosis (AE-IPF), clinically amyopathic dermatomyositis (CADM), epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI)-induced lung injury, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) infection named coronavirus disease 2019 (COVID-19). In this review, clinical information, physical findings, laboratory examinations, and findings on lung high-resolution computed tomography and lung histopathology are presented, focusing on majorly damaged cells in each disease. Furthermore, treatments that should be immediately initiated in clinical practice for each disease are illustrated to save patients with these diseases.
Collapse
Affiliation(s)
- Noriyuki Enomoto
- Second Division, Department of Internal Medicine, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan; ; Tel.: +81-53-435-2263; Fax: +81-53-435-2354
- Health Administration Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu 431-3192, Japan
| |
Collapse
|
117
|
Multiple Death Pathways of Neutrophils Regulate Alveolar Macrophage Proliferation. Cells 2022; 11:cells11223633. [PMID: 36429062 PMCID: PMC9688429 DOI: 10.3390/cells11223633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/19/2022] Open
Abstract
Alveolar macrophage (AM) proliferation and self-renewal play an important role in the lung tissue microenvironment. However, the impact of immune cells, especially the neutrophils, on AM homeostasis or function is not well characterized. In this study, we induced in vivo migration of neutrophils into bronchoalveolar lavage (BAL) fluid and lung using CXCL1, and then co-cultured these with AMs in vitro. Neutrophils in the BAL (BAL-neutrophils), rather than neutrophils of bone marrow (BM-neutrophils), were found to inhibit AM proliferation. Analysis of publicly available data showed high heterogeneity of lung neutrophils with distinct molecular signatures of BM- and blood-neutrophils. Unexpectedly, BAL-neutrophils from influenza virus PR8-infected mice (PR8-neutrophils) did not inhibit the proliferation of AMs. Bulk RNA sequencing further revealed that co-culture of AMs with PR8-neutrophils induced IFN-α and -γ responses and inflammatory response, and AMs co-cultured with BAL-neutrophils showed higher expression of metabolism- and ROS-associated genes; in addition, BAL-neutrophils from PR8-infected mice modulated AM polarization and phagocytosis. BAL-neutrophil-mediated suppression of AM proliferation was abrogated by a combination of inhibitors of different neutrophil death pathways. Collectively, our findings suggest that multiple cell death pathways of neutrophils regulate the proliferation of AMs. Targeting neutrophil death may represent a potential therapeutic strategy for improving AM homeostasis during respiratory diseases.
Collapse
|
118
|
Zhang Y, Li Y, Sun N, Tang H, Ye J, Liu Y, He Q, Fu Y, Zhu H, Jiang C, Xu J. NETosis is critical in patients with severe community-acquired pneumonia. Front Immunol 2022; 13:1051140. [PMID: 36466920 PMCID: PMC9709478 DOI: 10.3389/fimmu.2022.1051140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/24/2022] [Indexed: 11/12/2023] Open
Abstract
Pneumonia is the fourth leading cause of death globally, and the reason for the high mortality rate of patients with severe community-acquired pneumonia (SCAP) remains elusive. Corticosteroid treatment reduces mortality in adults with SCAP but can cause numerous adverse events. Therefore, novel therapeutic targets need to be explored and new adjunctive immune drugs are urgently required. We analyzed the transcriptome data of peripheral blood leukocytes from patients with SCAP and healthy controls from three perspectives: differentially expressed genes, predicted functions of differentially expressed long non-coding RNAs, and transcriptional read-through. We discovered that the NETosis pathway was top-ranked in patients with SCAP caused by diverse kinds of pathogens. This provides a potential therapeutic strategy for treating patients. Furthermore, we calculated the correlation between the expression of genes involved in NETosis and the ratio of arterial oxygen partial pressure to fractional inspired oxygen. We identified four novel potential therapeutic targets for NETosis in patients with SCAP, including H4C15, H3-5, DNASE1, and PRKCB. In addition, a higher occurrence of transcriptional read-through is associated with a worse outcome in patients with SCAP, which probably can explain the high mortality rate of patients with SCAP.
Collapse
Affiliation(s)
- Yiming Zhang
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan Li
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Na Sun
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hanqi Tang
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jun Ye
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yang Liu
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Quan He
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yangyang Fu
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huadong Zhu
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Chengyu Jiang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jun Xu
- Emergency Department, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
119
|
Hu Q, Zhang S, Yang Y, Yao JQ, Tang WF, Lyon CJ, Hu TY, Wan MH. Extracellular vesicles in the pathogenesis and treatment of acute lung injury. Mil Med Res 2022; 9:61. [PMID: 36316787 PMCID: PMC9623953 DOI: 10.1186/s40779-022-00417-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/19/2022] [Indexed: 11/05/2022] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are common life-threatening lung diseases associated with acute and severe inflammation. Both have high mortality rates, and despite decades of research on clinical ALI/ARDS, there are no effective therapeutic strategies. Disruption of alveolar-capillary barrier integrity or activation of inflammatory responses leads to lung inflammation and injury. Recently, studies on the role of extracellular vesicles (EVs) in regulating normal and pathophysiologic cell activities, including inflammation and injury responses, have attracted attention. Injured and dysfunctional cells often secrete EVs into serum or bronchoalveolar lavage fluid with altered cargoes, which can be used to diagnose and predict the development of ALI/ARDS. EVs secreted by mesenchymal stem cells can also attenuate inflammatory reactions associated with cell dysfunction and injury to preserve or restore cell function, and thereby promote cell proliferation and tissue regeneration. This review focuses on the roles of EVs in the pathogenesis of pulmonary inflammation, particularly ALI/ARDS.
Collapse
Affiliation(s)
- Qian Hu
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Shu Zhang
- Department of Emergency Medicine, Emergency Medical Laboratory, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yue Yang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Jia-Qi Yao
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Wen-Fu Tang
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Christopher J Lyon
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA.,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA
| | - Tony Ye Hu
- Center of Cellular and Molecular Diagnosis, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA. .,Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, 1430 Tulane Ave., New Orleans, LA, 70112, USA.
| | - Mei-Hua Wan
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital of Sichuan University, Chengdu, 610041, China. .,West China Hospital (Airport) of Sichuan University, Chengdu, 610299, China.
| |
Collapse
|
120
|
Latha K, Rao S, Sakamoto K, Watford WT. Tumor Progression Locus 2 Protects against Acute Respiratory Distress Syndrome in Influenza A Virus-Infected Mice. Microbiol Spectr 2022; 10:e0113622. [PMID: 35980186 PMCID: PMC9604045 DOI: 10.1128/spectrum.01136-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/16/2022] [Indexed: 12/30/2022] Open
Abstract
Excessive inflammation in patients with severe influenza disease may lead to acute lung injury that results in acute respiratory distress syndrome (ARDS). ARDS is associated with alveolar damage and pulmonary edema that severely impair gas exchange, leading to hypoxia. With no existing FDA-approved treatment for ARDS, it is important to understand the factors that lead to virus-induced ARDS development to improve prevention, diagnosis, and treatment. We have previously shown that mice deficient in the serine-threonine mitogen-activated protein kinase, Tpl2 (MAP3K8 or COT), succumb to infection with a typically low-pathogenicity strain of influenza A virus (IAV; HKX31, H3N2 [x31]). The goal of the current study was to evaluate influenza A virus-infected Tpl2-/- mice clinically and histopathologically to gain insight into the disease mechanism. We hypothesized that Tpl2-/- mice succumb to IAV infection due to development of ARDS-like disease and pulmonary dysfunction. We observed prominent signs of alveolar septal necrosis, hyaline membranes, pleuritis, edema, and higher lactate dehydrogenase (LDH) levels in the lungs of IAV-infected Tpl2-/- mice compared to wild-type (WT) mice from 7 to 9 days postinfection (dpi). Notably, WT mice showed signs of regenerating epithelium, indicative of repair and recovery, that were reduced in Tpl2-/- mice. Furthermore, biomarkers associated with human ARDS cases were upregulated in Tpl2-/- mice at 7 dpi, demonstrating an ARDS-like phenotype in Tpl2-/- mice in response to IAV infection. IMPORTANCE This study demonstrates the protective role of the serine-threonine mitogen-activated protein kinase, Tpl2, in influenza virus pathogenesis and reveals that host Tpl2 deficiency is sufficient to convert a low-pathogenicity influenza A virus infection into severe influenza disease that resembles ARDS, both histopathologically and transcriptionally. The IAV-infected Tpl2-/- mouse thereby represents a novel murine model for studying ARDS-like disease that could improve our understanding of this aggressive disease and assist in the design of better diagnostics and treatments.
Collapse
Affiliation(s)
- Krishna Latha
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Sanjana Rao
- Department of Genetics, University of Georgia, Athens, Georgia, USA
| | - Kaori Sakamoto
- Department of Pathology, University of Georgia, Athens, Georgia, USA
| | - Wendy T. Watford
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
121
|
Wan Y, Shen J, Ouyang J, Dong P, Hong Y, Liang L, Liu J. Bibliometric and visual analysis of neutrophil extracellular traps from 2004 to 2022. Front Immunol 2022; 13:1025861. [PMID: 36341351 PMCID: PMC9634160 DOI: 10.3389/fimmu.2022.1025861] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/10/2022] [Indexed: 12/27/2022] Open
Abstract
Background Neutrophil extracellular traps (NETs) are specialized structures formed by neutrophils that were initially found to be important in killing pathogenic bacteria during infection. With the development of related research, the relationship between NETs and diseases such as sepsis, cancer, and systemic lupus erythematosus has received close attention. However, there is a lack of reports that comprehensively and objectively present the current status of NETs-related studies. Therefore, this study aims to visually analyze the current status and trends of NETs-related research by means of bibliometrics and knowledge mapping. Methods NETs-related articles and reviews were retrieved using the Web of Science core collection subject search, and bibliometric analysis was performed in Excel 365, CiteSpace, VOSviewer, and Bibliometrix (R-Tool of R-Studio). Results A total of 4866 publications from 2004 to 2022 were included in the bibliometric analysis. The number of publications shows an increasing trend from year to year. Collaborative network analysis shows that the United States and Germany are the most influential countries in this field, with the highest number of publications and citations. The journal with the most publications is Frontiers in Immunology. Brinkmann Volker is an authoritative author in this field, and his publication "Neutrophil extracellular traps kill bacteria" is the most frequently cited. The literature and keyword analysis shows that the relationship between NETs and diseases (hematological diseases, sepsis, cancer, etc.) and cell death (apoptosis, necroptosis, pyroptosis, etc.) is a popular research topic. Currently, NETs and SARS-CoV-2-related studies are at the forefront of the field. Conclusion This study is the first to visualize the research in NETs-related fields using bibliometric methods, revealing the trends and frontiers of NETs research. This study will provide valuable references for scholars to find research focus questions and partners.
Collapse
Affiliation(s)
- Yantong Wan
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Junyi Shen
- The Second Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Jiafu Ouyang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Peng Dong
- College of Anesthesiology, Southern Medical University, Guangzhou, China
| | - Yinghao Hong
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lixin Liang
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jinghua Liu
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,*Correspondence: Jinghua Liu,
| |
Collapse
|
122
|
Hidalgo A, Libby P, Soehnlein O, Aramburu IV, Papayannopoulos V, Silvestre-Roig C. Neutrophil extracellular traps: from physiology to pathology. Cardiovasc Res 2022; 118:2737-2753. [PMID: 34648022 PMCID: PMC9586562 DOI: 10.1093/cvr/cvab329] [Citation(s) in RCA: 175] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/21/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
At the frontline of the host defence response, neutrophil antimicrobial functions have adapted to combat infections and injuries of different origins and magnitude. The release of web-like DNA structures named neutrophil extracellular traps (NETs) constitutes an important mechanism by which neutrophils prevent pathogen dissemination or deal with microorganisms of a bigger size. At the same time, nuclear and granule proteins with microbicidal activity bind to these DNA structures promoting the elimination of entrapped pathogens. However, these toxic properties may produce unwanted effects in the host, when neutrophils uncontrollably release NETs upon persistent inflammation. As a consequence, NET accumulation can produce vessel occlusion, tissue damage, and prolonged inflammation associated with the progression and exacerbation of multiple pathologic conditions. This review outlines recent advances in understanding the mechanisms of NET release and functions in sterile disease. We also discuss mechanisms of physiological regulation and the importance of neutrophil heterogeneity in NET formation and composition.
Collapse
Affiliation(s)
- Andres Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro 3, 28029, Madrid, Spain
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Oliver Soehnlein
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Straße 56, 48149, Münster, Germany
- Department of Physiology and Pharmacology (FyFa), Karolinska Institute, Solnavägen 1, 171 77, Stockholm, Sweden
| | - Iker Valle Aramburu
- Laboratory of Antimicrobial Defence, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Carlos Silvestre-Roig
- Institute for Experimental Pathology (ExPat), Center for Molecular Biology of Inflammation (ZMBE), University of Münster, Von-Esmarch-Straße 56, 48149, Münster, Germany
| |
Collapse
|
123
|
Pérez-Guerrero P, Illanes-Álvarez F, Márquez-Ruiz D, Campaña-Gómez I, Cuesta-Sancho S, Márquez-Coello M, Girón-González JA. Implication of Neutrophils Extracellular Traps in the Pathogenesis of SARS-CoV-2 pneumonia. Biomedicines 2022; 10:biomedicines10102638. [PMID: 36289900 PMCID: PMC9599188 DOI: 10.3390/biomedicines10102638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 12/15/2022] Open
Abstract
Peripheral blood polymorphonuclear neutrophils (PMNs) forming extracellular traps (NETs), as well as endothelial- and platelet-derived parameters, have been analyzed in patients with SARS-CoV-2 pneumonia, and their prognostic role has been evaluated. Eighty-seven consecutive patients hospitalized with SARS-CoV-2 pneumonia were prospectively selected. A sample of 30 healthy individuals served as the control group. Clinical and oxygenation (oxygen saturation to fraction of inspired oxygen ratio—SpO2/FiO2) characteristics and PMNs forming NETs, serum levels of myeloperoxidase, E-selectin, vascular cell adhesion molecule 1—VCAM1—vascular endothelial growth factor, P-selectin, platelet factor 4 and plasma concentrations of D-dimer were evaluated at hospital admission, at discharge and 14 days after discharge. Intensive care unit admission or death was the primary composite endpoint. Patients showed a higher number of PMNs forming NETs than healthy controls. The absolute number of PMNs forming NETs was inversely correlated with oxygen status (SpO2/FiO2) and positively with inflammatory (C-reactive protein, ferritin) markers and VCAM1. A decrease in, but not a normalization of NETs and endothelial-derived parameters was observed in patients who survived. In conclusion, the formation of NETs runs parallel to that of other inflammatory and endothelial activation markers, and is inverse to the oxygenation parameters, supporting a pathogenic role for PMNs in this entity.
Collapse
Affiliation(s)
- Patricia Pérez-Guerrero
- Servicio de Medicina Interna, Hospital Universitario Puerta del Mar, Facultad de Medicina, Universidad de Cádiz, Instituto para la Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - Francisco Illanes-Álvarez
- Servicio de Medicina Interna, Hospital Universitario Puerta del Mar, Facultad de Medicina, Universidad de Cádiz, Instituto para la Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - Denisse Márquez-Ruiz
- Servicio de Medicina Interna, Hospital Universitario Puerta del Mar, Facultad de Medicina, Universidad de Cádiz, Instituto para la Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - Irene Campaña-Gómez
- Servicio de Medicina Interna, Hospital Universitario Puerta del Mar, Facultad de Medicina, Universidad de Cádiz, Instituto para la Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - Sara Cuesta-Sancho
- Departamento de Inmunología, Facultad de Medicina, Universidad de Valladolid, 47003 Valladolid, Spain
| | - Mercedes Márquez-Coello
- Servicio de Medicina Interna, Hospital Universitario Puerta del Mar, Facultad de Medicina, Universidad de Cádiz, Instituto para la Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
| | - José-Antonio Girón-González
- Servicio de Medicina Interna, Hospital Universitario Puerta del Mar, Facultad de Medicina, Universidad de Cádiz, Instituto para la Investigación e Innovación Biomédica de Cádiz (INiBICA), 11009 Cádiz, Spain
- Correspondence:
| |
Collapse
|
124
|
Jung SH, Lee KT. Atherosclerosis by Virus Infection—A Short Review. Biomedicines 2022; 10:biomedicines10102634. [PMID: 36289895 PMCID: PMC9599298 DOI: 10.3390/biomedicines10102634] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis manifests by the thickening of artery walls and their narrowed channels through the accumulation of plaque. It is one of the most important indicators of cardiovascular disease. It can be caused by various factors, such as smoking, a high cholesterol diet, hypertension, hyperglycemia, and genetic factors. However, atherosclerosis can also develop due to infection. It has been reported that some bacteria and viruses can cause the development of atherosclerosis. Examples of these viruses are influenza viruses, herpes viruses, hepatitis viruses, or papillomaviruses, which are all prevalent and eminent globally for infecting the population worldwide. Moreover, many patients with coronavirus disease 2019 (COVID-19) showed symptoms of cardiovascular disease. In this review paper, the viruses linked to the development of atherosclerosis are introduced, and their viral characteristics, the mechanisms of the development of atherosclerosis, and the current vaccines and antiviral treatment methods are summarized.
Collapse
Affiliation(s)
- Seang-Hwan Jung
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02247, Korea
- Correspondence: (S.-H.J.); (K.-T.L.)
| | - Kyung-Tae Lee
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02247, Korea
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02247, Korea
- Correspondence: (S.-H.J.); (K.-T.L.)
| |
Collapse
|
125
|
Chen L, Hua J, He X. Co-expression network analysis identifies potential candidate hub genes in severe influenza patients needing invasive mechanical ventilation. BMC Genomics 2022; 23:703. [PMID: 36243706 PMCID: PMC9569050 DOI: 10.1186/s12864-022-08915-9] [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: 06/21/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Influenza is a contagious disease that affects people of all ages and is linked to considerable mortality during epidemics and occasional outbreaks. Moreover, effective immunological biomarkers are needed for elucidating aetiology and preventing and treating severe influenza. Herein, we aimed to evaluate the key genes linked with the disease severity in influenza patients needing invasive mechanical ventilation (IMV). Three gene microarray data sets (GSE101702, GSE21802, and GSE111368) from blood samples of influenza patients were made available by the Gene Expression Omnibus (GEO) database. The GSE101702 and GSE21802 data sets were combined to create the training set. Hub indicators for IMV patients with severe influenza were determined using differential expression analysis and Weighted correlation network analysis (WGCNA) from the training set. The receiver operating characteristic curve (ROC) was also used to evaluate the hub genes from the test set's diagnostic accuracy. Different immune cells' infiltration levels in the expression profile and their correlation with hub gene markers were examined using single-sample gene set enrichment analysis (ssGSEA). RESULTS In the present study, we evaluated a total of 447 differential genes. WGCNA identified eight co-expression modules, with the red module having the strongest correlation with IMV patients. Differential genes were combined to obtain 3 hub genes (HLA-DPA1, HLA-DRB3, and CECR1). The identified genes were investigated as potential indicators for patients with severe influenza who required IMV using the least absolute shrinkage and selection operator (LASSO) approach. The ROC showed the diagnostic value of the three hub genes in determining the severity of influenza. Using ssGSEA, it has been revealed that the expression of key genes was negatively correlated with neutrophil activation and positively associated with adaptive cellular immune response. CONCLUSION We evaluated three novel hub genes that could be linked to the immunopathological mechanism of severe influenza patients who require IMV treatment and could be used as potential biomarkers for severe influenza prevention and treatment.
Collapse
Affiliation(s)
- Liang Chen
- Department of Infectious Diseases, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Jie Hua
- Department of Gastroenterology, Liyang People's Hospital, Liyang Branch Hospital of Jiangsu Province Hospital, Nanjing, China
| | - Xiaopu He
- Department of Geriatric Gastroenterology, The First Affiliated Hospital With Nanjing Medical University, No.300 Guangzhou Road, Nanjing city, 210029, Jiangsu Province, China.
| |
Collapse
|
126
|
Zhang Z, Gao Y, Li L, Luo J, Gao R. Deficiency of C-reactive protein or human C-reactive protein transgenic treatment aggravates influenza A infection in mice. Front Immunol 2022; 13:1028458. [PMID: 36275680 PMCID: PMC9584053 DOI: 10.3389/fimmu.2022.1028458] [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: 08/26/2022] [Accepted: 09/20/2022] [Indexed: 12/15/2022] Open
Abstract
C-reactive protein (CRP) has been shown to be a potential candidate target in the immunotherapy of severe influenza A infection. However, it is unclear on the pathogenesis associated with CRP in influenza infections. Here, we used influenza A H1N1 CA04 to infect human CRP transgenic mice (KI), CRP knockout mice (KO), and wild-type mice (WT), respectively, and compared the viral pathogenicity and associated immune response in those mice. The results showed that CA04 infection resulted in 100%, 80%, and 60% death in KO, KI, and WT mice, respectively. Compared to WT mice, CA04 infection resulted in higher TCID50 in lungs on day 3 after infection but lowered HI antibody titers in sera of survivors on day 21 after infection in KI mice. ELISA assay showed that IFN-γ concentration was significantly increased in sera of WT, KI, or KO mice on day 7 after infection, and IL-17 was remarkably increased in sera of WT mice but decreased in sera of KI mice while no significant change in sera of KO mice on day 3 or 7 after infection. Quantitative RT-PCR showed that the relative expression levels of immune checkpoint CTLA-4, LAIR-1, GITR, BTLA, TIM-3, or PD-1 mRNA in the lung presented decreased levels on day 3 or 7 after infection in KI or KO mice. The correlation analysis showed that mRNA expression levels of the 6 molecules positively correlated with viral TICD50 in WT mice but negatively correlated with viral TCID50 in KI or KO mice. However, only LAIR-1 presented a significant correlation in each lung tissue of WT, KI, or KO mice with CA07 infection statistically. IHC results showed that LAIR-1 positive cells could be found in WT, KO, or KI mice lung tissues with CA04 infection, and the positive cells were mainly distributed in an inflammatory dense area. Our results suggested that deficiency of CRP or human CRP transgenic treatment aggravates influenza A virus infection in mice. CRP is a double sword in immune regulation of influenza infection in which IL-17 and immune checkpoint may be involved.
Collapse
Affiliation(s)
- Zhuohan Zhang
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yongjun Gao
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li Li
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Junhao Luo
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Rongbao Gao
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- National Health Commission of People's Republic of China (NHC) Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- *Correspondence: Rongbao Gao,
| |
Collapse
|
127
|
Carstensen S, Müller M, Tan GLA, Pasion KA, Hohlfeld JM, Herrera VLM, Ruiz-Opazo N. “Rogue” neutrophil-subset [DEspR+CD11b+/CD66b+] immunotype is an actionable therapeutic target for neutrophilic inflammation-mediated tissue injury – studies in human, macaque and rat LPS-inflammation models. Front Immunol 2022; 13:1008390. [PMID: 36275710 PMCID: PMC9581391 DOI: 10.3389/fimmu.2022.1008390] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background and objective The correlation (Rs > 0.7) of neutrophils expressing the dual endothelin1/signal peptide receptor (DEspR+CD11b+/CD66b+) with severity of hypoxemia (SF-ratio) and multi-organ failure (SOFA-score) in patients with acute respiratory distress syndrome (ARDS) suggest the hypothesis that the DEspR+ neutrophil-subset is an actionable therapeutic target in ARDS. To test this hypothesis, we conducted in vivo studies to validate DEspR+ neutrophil-subset as therapeutic target and test efficacy of DEspR-inhibition in acute neutrophilic hyperinflammation models. Methods We performed tests in lipopolysaccharide (LPS)-induced acute neutrophilic inflammation in three species – human, rhesus macaque, rat – with increasing dose-dependent severity. We measured DEspR+CD66b+ neutrophils in bronchoalveolar lavage fluid (BALF) in healthy volunteers (HVs) 24-hours after segmental LPS-challenge by ChipCytometry, and DEspR+CD11b+ neutrophils in whole blood and BALF in an LPS-induced transient acute lung injury (ALI) model in macaques. We determined anti-DEspR antibody efficacy in vivo in LPS-ALI macaque model and in high-mortality LPS-induced encephalopathy in hypertensive rats. Results ChipCytometry detected increased BALF total neutrophil and DEspR+CD66b+ neutrophil counts after segmental LPS-challenge compared to baseline (P =0.034), as well as increased peripheral neutrophil counts and neutrophil-lymphocyte ratio (NLR) compared to pre-LPS level (P <0.05). In the LPS-ALI macaque model, flow cytometry detected increased DEspR+ and DEspR[-] neutrophils in BALF, which was associated with moderate-severe hypoxemia. After determining pharmacokinetics of single-dose anti-DEspR[hu6g8] antibody, one-time pre-LPS anti-DEspR treatment reduced hypoxemia (P =0.03) and neutrophil influx into BALF (P =0.0001) in LPS-ALI vs vehicle mock-treated LPS-ALI macaques. Ex vivo live cell imaging of macaque neutrophils detected greater “intrinsic adhesion to hard-surface” in DEspR+ vs DEspR[-] neutrophils (P <0.001). Anti-DEspR[hu6g8] antibody abrogated intrinsic high adhesion in DEspR+ neutrophils, but not in DEspR[-] neutrophils (P <0.001). In the LPS-encephalopathy rat model, anti-DEspR[10a3] antibody treatment increased median survival (P =0.0007) and exhibited brain target engagement and bioeffects. Conclusion Detection of increased DEspR+ neutrophil-subset in human BALF after segmental LPS-challenge supports the correlation of circulating DEspR+ neutrophil counts with severity measure (SOFA-score) in ARDS. Efficacy and safety of targeted inhibition of DEspR+CD11b+ neutrophil-subset in LPS-induced transient-ALI and high-mortality encephalopathy models identify a potential therapeutic target for neutrophil-mediated secondary tissue injury.
Collapse
Affiliation(s)
- Saskia Carstensen
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Department of Biomarker Analysis and Development, Hannover, Germany
| | - Meike Müller
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Department of Biomarker Analysis and Development, Hannover, Germany
| | - Glaiza L. A. Tan
- Whitaker Cardiovascular Institute and Department of Medicine, Boston University School of Medicine, Boston MA, United States
| | - Khristine Amber Pasion
- Whitaker Cardiovascular Institute and Department of Medicine, Boston University School of Medicine, Boston MA, United States
| | - Jens M. Hohlfeld
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Department of Biomarker Analysis and Development, Hannover, Germany
- Department of Respiratory Medicine, Hannover Medical School, Hannover, Germany
- Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Victoria L. M. Herrera
- Whitaker Cardiovascular Institute and Department of Medicine, Boston University School of Medicine, Boston MA, United States
| | - Nelson Ruiz-Opazo
- Whitaker Cardiovascular Institute and Department of Medicine, Boston University School of Medicine, Boston MA, United States
- *Correspondence: Nelson Ruiz-Opazo,
| |
Collapse
|
128
|
Agarwal S. Neutrophil-Lymphocyte Ratio Predicting Case Severity in SARS-CoV-2 Infection: A Review. Cureus 2022; 14:e29760. [PMID: 36187170 PMCID: PMC9521818 DOI: 10.7759/cureus.29760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2022] [Indexed: 12/15/2022] Open
|
129
|
Hastak PS, Andersen CR, Kelleher AD, Sasson SC. Frontline workers: Mediators of mucosal immunity in community acquired pneumonia and COVID-19. Front Immunol 2022; 13:983550. [PMID: 36211412 PMCID: PMC9539803 DOI: 10.3389/fimmu.2022.983550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
The current COVID-19 pandemic has highlighted a need to further understand lung mucosal immunity to reduce the burden of community acquired pneumonia, including that caused by the SARS-CoV-2 virus. Local mucosal immunity provides the first line of defence against respiratory pathogens, however very little is known about the mechanisms involved, with a majority of literature on respiratory infections based on the examination of peripheral blood. The mortality for severe community acquired pneumonia has been rising annually, even prior to the current pandemic, highlighting a significant need to increase knowledge, understanding and research in this field. In this review we profile key mediators of lung mucosal immunity, the dysfunction that occurs in the diseased lung microenvironment including the imbalance of inflammatory mediators and dysbiosis of the local microbiome. A greater understanding of lung tissue-based immunity may lead to improved diagnostic and prognostic procedures and novel treatment strategies aimed at reducing the disease burden of community acquired pneumonia, avoiding the systemic manifestations of infection and excess morbidity and mortality.
Collapse
Affiliation(s)
- Priyanka S. Hastak
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| | - Christopher R. Andersen
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
- Intensive Care Unit, Royal North Shore Hospital, Sydney, NSW, Australia
- Critical Care and Trauma Division, The George Institute for Global Health, Sydney, NSW, Australia
| | - Anthony D. Kelleher
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| | - Sarah C. Sasson
- The Kirby Institute, Immunovirology and Pathogenesis Program, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
130
|
Sung PS, Peng YC, Yang SP, Chiu CH, Hsieh SL. CLEC5A is critical in Pseudomonas aeruginosa-induced acute lung injury. JCI Insight 2022; 7:156613. [PMID: 36048544 PMCID: PMC9676025 DOI: 10.1172/jci.insight.156613] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
Pseudomonas aeruginosa is one of the most common nosocomial infections worldwide, and it frequently causes ventilator-associated acute pneumonia in immunocompromised patients. Abundant neutrophil extracellular traps (NETs) contribute to acute lung injury, thereby aggravating ventilator-induced lung damage. While pattern recognition receptors (PRRs) TLR4 and TLR5 are required for host defense against P. aeruginosa invasion, the PRR responsible for P. aeruginosa–induced NET formation, proinflammatory cytokine release, and acute lung injury remains unclear. We found that myeloid C-type lectin domain family 5 member A (CLEC5A) interacts with LPS of P. aeruginosa and is responsible for P. aeruginosa–induced NET formation and lung inflammation. P. aeruginosa activates CLEC5A to induce caspase-1–dependent NET formation, but it neither causes gasdermin D (GSDMD) cleavage nor contributes to P. aeruginosa–induced neutrophil death. Blockade of CLEC5A attenuates P. aeruginosa–induced NETosis and lung injury, and simultaneous administration of anti-CLEC5A mAb with ciprofloxacin increases survival rate and decreases collagen deposition in the lungs of mice challenged with a lethal dose of P. aeruginosa. Thus, CLEC5A is a promising therapeutic target to reduce ventilator-associated lung injury and fibrosis in P. aeruginosa–induced pneumonia.
Collapse
Affiliation(s)
- Pei-Shan Sung
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chun Peng
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shao-Ping Yang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Cheng-Hsun Chiu
- Department of Pediatrics, Chang Gung Children's Hospital, Taoyuan, Taiwan
| | | |
Collapse
|
131
|
Effects and Action Mechanism of Huoxue Tongluo Formula on the Formation of Neutrophil Extracellular Traps. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1240967. [PMID: 36034958 PMCID: PMC9410787 DOI: 10.1155/2022/1240967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 12/03/2022]
Abstract
Excessive infiltration and uncontrolled activation of neutrophil extracellular traps (NETs) are likely to destroy normal tissue architecture and cause uncontrolled inflammation. The present research attempted to screen potential signaling pathways of Huoxue Tongluo Formula (HXTLF) affecting the formation of NETs using network pharmacology technique. Active chemical components of HXTLF and therapeutic targets related to vasculitis were screened, and a chemical components-targets network diagram of HXTLF was constructed by Cytoscape. Finally, the inhibitory effect and mechanism of HXTLF on the formation of NETs were explored in vitro using LPS-induced NETs. Immunofluorescence and Western blot were conducted to determine the protein fluorescence intensity and relative expression. The experimental results illustrated that HXTLF mediated the expression levels of H3Cit and myeloperoxidase (MPO) protein in neutrophils activated by LPS, inhibited NETs formation, and reduced the concentration of interleukin- (IL-) 1β, a proinflammatory factor in cells. Additionally, we activated and inhibited the AKT1 signaling pathway using the corresponding activator and inhibitor to explore the regulatory mechanism of HXTLF on AKT1 and other molecules in the treatment of vasculitis. The results demonstrated that HXTLF could inhibit the phosphorylation of AKT1, IKK, and NF-κB proteins, inhibit NETs formation, and reduce IL-1β concentration, indicating that AKT1 exerts a vital role in the treatment of vasculitis after HXTLF administration. The current study initially revealed the pharmacological mechanism of HXTLF for vasculitis management using network pharmacology techniques and tests in vitro, which is expected to provide important theoretical basis for elucidating the molecular mechanism of HXTLF and promoting its clinical application.
Collapse
|
132
|
Long G, Gong R, Wang Q, Zhang D, Huang C. Role of released mitochondrial DNA in acute lung injury. Front Immunol 2022; 13:973089. [PMID: 36059472 PMCID: PMC9433898 DOI: 10.3389/fimmu.2022.973089] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 08/01/2022] [Indexed: 12/02/2022] Open
Abstract
Acute lung injury(ALI)/acute respiratory distress syndrome(ARDS) is a form of acute-onset hypoxemic respiratory failure characterised by an acute, diffuse, inflammatory lung injury, and increased alveolar-capillary permeability, which is caused by a variety of pulmonary or nonpulmonary insults. Recently, aberrant mitochondria and mitochondrial DNA(mtDNA) level are associated with the development of ALI/ARDS, and plasma mtDNA level shows the potential to be a promising biomarker for clinical diagnosis and evaluation of lung injury severity. In mechanism, the mtDNA and its oxidised form, which are released from impaired mitochondria, play a crucial role in the inflammatory response and histopathological changes in the lung. In this review, we discuss mitochondrial outer membrane permeabilisation (MOMP), mitochondrial permeability transition pore(mPTP), extracellular vesicles (EVs), extracellular traps (ETs), and passive release as the principal mechanisms for the release of mitochondrial DNA into the cytoplasm and extracellular compartments respectively. Further, we explain how the released mtDNA and its oxidised form can induce inflammatory cytokine production and aggravate lung injury through the Toll-like receptor 9(TLR9) signalling, cytosolic cGAS-stimulator of interferon genes (STING) signalling (cGAS-STING) pathway, and inflammasomes activation. Additionally, we propose targeting mtDNA-mediated inflammatory pathways as a novel therapeutic approach for treating ALI/ARDS.
Collapse
Affiliation(s)
- Gangyu Long
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Rui Gong
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qian Wang
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Dingyu Zhang
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, China
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Dingyu Zhang, ; Chaolin Huang,
| | - Chaolin Huang
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Research Center for Infectious Diseases, Wuhan, China
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Wuhan, China
- Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan, China
- *Correspondence: Dingyu Zhang, ; Chaolin Huang,
| |
Collapse
|
133
|
Ouyang Y, Liu J, Wen S, Xu Y, Zhang Z, Pi Y, Chen D, Su Z, Liang Z, Wang Y, Guo L. Association between chronic obstructive pulmonary disease and periodontitis: The common role of innate immune cells? Cytokine 2022; 158:155982. [PMID: 35932499 DOI: 10.1016/j.cyto.2022.155982] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 07/14/2022] [Accepted: 07/25/2022] [Indexed: 11/19/2022]
Abstract
Innate immune cells are of broad interest in a variety of diseases. These cells include neutrophils, macrophages, dendritic cells and mast cells, etc. Innate immune cells are often mentioned in inflammatory diseases as the first line of defense against pathogens' invasion. As chronic obstructive pulmonary disease and periodontitis are inflammatory diseases, innate immune cells play an important role in the development of both diseases. COPD and periodontitis are common epidemic diseases with a very high prevalence, thus affecting a large number of people and also reducing the quality of life of patients. In addition, epidemiological studies suggested a link between the two, creating a co-morbid burden, but the mechanism of the link is yet to be explained. This article discusses the possible mechanism of the link between the two diseases in terms of innate immune cells and discusses possible future targeted therapies that could alleviate the burden on patients.
Collapse
Affiliation(s)
- Yuanting Ouyang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Jiaohong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Siyi Wen
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Yixin Xu
- Department of Orthodontic, Stomatological Hospital, Southern Medical University, China
| | - Zhiyi Zhang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Yixing Pi
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Ding Chen
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Zhikang Su
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Zitian Liang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China
| | - Yan Wang
- Guangzhou Medical University, Guangzhou, Guangdong, China; Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong, China.
| | - Lvhua Guo
- Guangzhou Medical University, Guangzhou, Guangdong, China; Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
134
|
McGrath JJC, Vanderstocken G, Dvorkin-Gheva A, Cass SP, Afkhami S, Fantauzzi MF, Thayaparan D, Reihani A, Wang P, Beaulieu A, Shen P, Morissette M, Jiménez-Saiz R, Revill SD, Tabuchi A, Zabini D, Lee WL, Richards CD, Miller MS, Ask K, Kuebler WM, Simpson JA, Stämpfli MR. Cigarette smoke augments CSF3 expression in neutrophils to compromise alveolar-capillary barrier function during influenza infection. Eur Respir J 2022; 60:2102049. [PMID: 35058252 DOI: 10.1183/13993003.02049-2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 12/29/2021] [Indexed: 11/05/2022]
Abstract
BACKGROUND Cigarette smokers are at increased risk of acquiring influenza, developing severe disease and requiring hospitalisation/intensive care unit admission following infection. However, immune mechanisms underlying this predisposition are incompletely understood, and therapeutic strategies for influenza are limited. METHODS We used a mouse model of concurrent cigarette smoke exposure and H1N1 influenza infection, colony-stimulating factor (CSF)3 supplementation/receptor (CSF3R) blockade and single-cell RNA sequencing (scRNAseq) to investigate this relationship. RESULTS Cigarette smoke exposure exacerbated features of viral pneumonia such as oedema, hypoxaemia and pulmonary neutrophilia. Smoke-exposed infected mice demonstrated an increase in viral (v)RNA, but not replication-competent viral particles, relative to infection-only controls. Interstitial rather than airspace neutrophilia positively predicted morbidity in smoke-exposed infected mice. Screening of pulmonary cytokines using a novel dysregulation score identified an exacerbated expression of CSF3 and interleukin-6 in the context of smoke exposure and influenza. Recombinant (r)CSF3 supplementation during influenza aggravated morbidity, hypothermia and oedema, while anti-CSF3R treatment of smoke-exposed infected mice improved alveolar-capillary barrier function. scRNAseq delineated a shift in the distribution of Csf3 + cells towards neutrophils in the context of cigarette smoke and influenza. However, although smoke-exposed lungs were enriched for infected, highly activated neutrophils, gene signatures of these cells largely reflected an exacerbated form of typical influenza with select unique regulatory features. CONCLUSION This work provides novel insight into the mechanisms by which cigarette smoke exacerbates influenza infection, unveiling potential therapeutic targets (e.g. excess vRNA accumulation, oedematous CSF3R signalling) for use in this context, and potential limitations for clinical rCSF3 therapy during viral infectious disease.
Collapse
Affiliation(s)
- Joshua J C McGrath
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
- Authors contributed equally
| | - Gilles Vanderstocken
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
- Authors contributed equally
| | - Anna Dvorkin-Gheva
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Steven P Cass
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Sam Afkhami
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew F Fantauzzi
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Danya Thayaparan
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amir Reihani
- Firestone Institute for Respiratory Health, St Joseph's Healthcare Hamilton, Hamilton, ON, Canada
- The Research Institute of St Joe's Hamilton, Hamilton, ON, Canada
| | - Peiyao Wang
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Ashley Beaulieu
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Pamela Shen
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Mathieu Morissette
- Dept of Medicine, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Université Laval, Quebec City, QC, Canada
| | - Rodrigo Jiménez-Saiz
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
- Dept of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa, Madrid, Spain
- Dept of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Spencer D Revill
- Firestone Institute for Respiratory Health, St Joseph's Healthcare Hamilton, Hamilton, ON, Canada
- The Research Institute of St Joe's Hamilton, Hamilton, ON, Canada
| | - Arata Tabuchi
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, Canada
| | - Diana Zabini
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, Canada
| | - Warren L Lee
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, Canada
| | - Carl D Richards
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
- Firestone Institute for Respiratory Health, St Joseph's Healthcare Hamilton, Hamilton, ON, Canada
| | - Wolfgang M Kuebler
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, Canada
- Institute of Physiology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Jeremy A Simpson
- Dept of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Martin R Stämpfli
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
- Dept of Medicine, McMaster University, Hamilton, ON, Canada
- Firestone Institute for Respiratory Health, St Joseph's Healthcare Hamilton, Hamilton, ON, Canada
| |
Collapse
|
135
|
Zamani Rarani F, Zamani Rarani M, Hamblin MR, Rashidi B, Hashemian SMR, Mirzaei H. Comprehensive overview of COVID-19-related respiratory failure: focus on cellular interactions. Cell Mol Biol Lett 2022; 27:63. [PMID: 35907817 PMCID: PMC9338538 DOI: 10.1186/s11658-022-00363-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 07/06/2022] [Indexed: 01/08/2023] Open
Abstract
The pandemic outbreak of coronavirus disease 2019 (COVID-19) has created health challenges in all parts of the world. Understanding the entry mechanism of this virus into host cells is essential for effective treatment of COVID-19 disease. This virus can bind to various cell surface molecules or receptors, such as angiotensin-converting enzyme 2 (ACE2), to gain cell entry. Respiratory failure and pulmonary edema are the most important causes of mortality from COVID-19 infections. Cytokines, especially proinflammatory cytokines, are the main mediators of these complications. For normal respiratory function, a healthy air-blood barrier and sufficient blood flow to the lungs are required. In this review, we first discuss airway epithelial cells, airway stem cells, and the expression of COVID-19 receptors in the airway epithelium. Then, we discuss the suggested molecular mechanisms of endothelial dysfunction and blood vessel damage in COVID-19. Coagulopathy can be caused by platelet activation leading to clots, which restrict blood flow to the lungs and lead to respiratory failure. Finally, we present an overview of the effects of immune and non-immune cells and cytokines in COVID-19-related respiratory failure.
Collapse
Affiliation(s)
- Fahimeh Zamani Rarani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Zamani Rarani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028 South Africa
| | - Bahman Rashidi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Mohammad Reza Hashemian
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Mirzaei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, IR Iran
| |
Collapse
|
136
|
Le A, Liu W, Wu C, Hu P, Zou J, Wu Y, Kuang L. Polymorphonuclear neutrophil activation by Src phosphorylation contributes to HLA-A2 antibody-induced transfusion-related acute lung injury. Mol Immunol 2022; 150:9-19. [PMID: 35914412 DOI: 10.1016/j.molimm.2022.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/18/2022] [Accepted: 04/21/2022] [Indexed: 11/25/2022]
Abstract
Human leukocyte antigen (HLA)-A2 antibody contributes to the pathogenesis of transfusion-related acute lung injury (TRALI) via polymorphonuclear neutrophil (PMN) activation, but the signaling pathways involved this process remain largely undefined. In this study, we sought to study the signaling pathways involved in the pathogenesis of HLA-A2-induced TRALI. Lipopolysaccharide (LPS), and the plasma from the HLA-A2 antibody-positive donors were utilized to establish a rat model of TRALI. Human pulmonary endothelial cells (HPMECs) were in vitro co-cultured with HLA-A2 antibody-treated PMNs and then treated with LPS to induce a cytotoxicity model. The effects of HLA-A2 antibody on HPMEC injury were evaluated in this model. Besides, dasatinib was used to block the Src phosphorylation to explore whether Src involved in the TRALI or HPMEC injury induced by HLA-A2 antibody. The HLA-A2 antibody plus LPS induced TRALI and stimulated PMN activation in rats. HLA-A2 antibody-induced TRALI could be attenuated via depletion of PMN. HLA-A2 antibody activated NF-κB and NLRP3 inflammasome. In addition, HLA-A2 antibody aggravated the HPMEC injuries and the release of PMN surfaces makers, but dasatinib treatment reversed this effect, indicating that HLA-A2 antibody activated PMNs and exacerbated TRALI by stimulating phosphorylation of Src followed by activation of NF-κB and NLRP3 inflammasome, which was validated in vivo. In summary, HLA-A2 induced PMNs by activating NF-κB/NLRP3 inflammasome via phosphorylated-Src elevation, thereby exacerbating TRALI. This study highlights promising target for the treatment of antibody-mediated TRALI.
Collapse
Affiliation(s)
- Aiping Le
- Departments of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China.
| | - Wei Liu
- Departments of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Chenggao Wu
- Departments of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Piaoping Hu
- Departments of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Juan Zou
- Departments of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Yize Wu
- Departments of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| | - Linju Kuang
- Departments of Blood Transfusion, The First Affiliated Hospital of Nanchang University, Nanchang 330006, PR China
| |
Collapse
|
137
|
Wu Q, Tu H, Li J. Multifaceted Roles of Chemokine C-X-C Motif Ligand 7 in Inflammatory Diseases and Cancer. Front Pharmacol 2022; 13:914730. [PMID: 35837284 PMCID: PMC9273993 DOI: 10.3389/fphar.2022.914730] [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: 04/07/2022] [Accepted: 06/07/2022] [Indexed: 11/13/2022] Open
Abstract
Over recent years, C-X-C motif ligand 7 (CXCL7) has received widespread attention as a chemokine involved in inflammatory responses. Abnormal production of the chemokine CXCL7 has been identified in different inflammatory diseases; nevertheless, the exact role of CXCL7 in the pathogenesis of inflammatory diseases is not fully understood. Persistent infection or chronic inflammation can induce tumorigenesis and progression. Previous studies have shown that the pro-inflammatory chemokine CXCL7 is also expressed by malignant tumor cells and that binding of CXCL7 to its cognate receptors C-X-C chemokine receptor 1 (CXCR1) and C-X-C chemokine receptor 2 (CXCR2) can influence tumor biological behavior (proliferation, invasion, metastasis, and tumor angiogenesis) in an autocrine and paracrine manner. CXCL7 and its receptor CXCR1/CXCR2, which are aberrantly expressed in tumors, may represent new targets for clinical tumor immunotherapy.
Collapse
Affiliation(s)
- Qianmiao Wu
- Department of Hematology, Second Affiliated Hospital of Nanchang University, Nanchang, China.,Department of Medicine, Nanchang University, Nanchang, China
| | - Huaijun Tu
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jian Li
- Department of Hematology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
138
|
Xiao S, Liu L, Sun Z, Liu X, Xu J, Guo Z, Yin X, Liao F, Xu J, You Y, Zhang T. Network Pharmacology and Experimental Validation to Explore the Mechanism of Qing-Jin-Hua-Tan-Decoction Against Acute Lung Injury. Front Pharmacol 2022; 13:891889. [PMID: 35873580 PMCID: PMC9304690 DOI: 10.3389/fphar.2022.891889] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/24/2022] [Indexed: 11/29/2022] Open
Abstract
Qing-Jin-Hua-Tan-Decoction (QJHTD), a classic famous Chinese ancient prescription, has been used for treatment of pulmonary diseases since Ming Dynasty. A total of 22 prototype compounds of QJHTD absorbed into rat blood were chosen as candidates for the pharmacological network analysis and molecular docking. The targets from the intersection of compound target and ALI disease targets were used for GO and KEGG enrichment analyses. Molecular docking was adopted to further verify the interactions between 22 components and the top 20 targets with higher degree values in the component-target-pathway network. In vitro experiments were performed to verify the results of network pharmacology using SPR experiments, Western blot experiments, and the PMA-induced neutrophils to produce neutrophil extracellular trap (NET) model. The compound-target-pathway network includes 176 targets and 20 signaling pathways in which the degree of MAPK14, CDK2, EGFR, F2, SRC, and AKT1 is higher than that of other targets and which may be potential disease targets. The biological processes in QJHTD for ALI mainly included protein phosphorylation, response to wounding, response to bacterium, regulation of inflammatory response, and so on. KEGG enrichment analyses revealed multiple signaling pathways, including lipid and atherosclerosis, HIF-1 signaling pathway, renin-angiotensin system, and neutrophil extracellular trap formation. The molecular docking results showed that baicalin, oroxylin A-7-glucuronide, hispidulin-7-O-β-D-glucuronide, wogonoside, baicalein, wogonin, tianshic acid, and mangiferin can be combined with most of the targets, which might be the core components of QJHTD in treatment of ALI. Direct binding ability of baicalein, wogonin, and baicalin to thrombin protein was all micromolar, and their KD values were 11.92 μM, 1.303 μM, and 1.146 μM, respectively, revealed by SPR experiments, and QJHTD could inhibit Src phosphorylation in LPS-activated neutrophils by Western blot experiments. The experimental results of PMA-induced neutrophils to produce NETs indicated that QJHTD could inhibit the production of NETs. This study revealed the active compounds, effective targets, and potential pharmacological mechanisms of QJHTD acting on ALI.
Collapse
Affiliation(s)
- Shunli Xiao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhengxiao Sun
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoqian Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhongyuan Guo
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xiaojie Yin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fulong Liao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jun Xu
- National and Local United Engineering Laboratory of Modern Preparation and Quality Control Technology of Traditional Chinese Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| | - Yun You
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tiejun Zhang
- National and Local United Engineering Laboratory of Modern Preparation and Quality Control Technology of Traditional Chinese Medicine, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| |
Collapse
|
139
|
Casting a wide NET: an update on uncontrolled NETosis in response to COVID-19 infection. Clin Sci (Lond) 2022; 136:1047-1052. [PMID: 35791847 PMCID: PMC9264284 DOI: 10.1042/cs20220039] [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: 05/08/2022] [Revised: 06/13/2022] [Accepted: 06/17/2022] [Indexed: 12/15/2022]
Abstract
Abstract
Dysregulation of neutrophil extracellular trap (NET) formation has been shown to mediate disease pathology in multiple viral infections, including SARS-CoV-2. At the beginning of COVID-19 pandemic, Thierry and Roch wrote a perspective on the mechanisms by which severe SARS-CoV-2 infection may lead to uncontrolled NET formation that leads to acute respiratory distress syndrome (ARDS), systemic vascular permeability, and end organ damage. In this commentary, the progress that has been made in regards to the ideas postulated by the perspective will be discussed, with a focus on the therapeutics that target NET formation.
Collapse
|
140
|
Cui X, Liang L, Geng H, Liu Y, Xi J, Wang J, Ching TB, Bee EG, Chai Y, Wu S, Jin D, Xie Y. Efficacy, Safety and Mechanism of Jinzhen Oral Liquid in the Treatment of Acute Bronchitis in Children: A Randomized, Double-Blind, Multicenter Clinical Trial Protocol. Front Pharmacol 2022; 13:948236. [PMID: 35847029 PMCID: PMC9283571 DOI: 10.3389/fphar.2022.948236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Acute bronchitis (AB) is a common disease in pediatrics. Prolonged AB may develop into chronic bronchitis. Bronchitis caused by the influenza virus can lead to severe hypoxia or insufficient ventilation, causing great harm to patients and increasing the burden on children and society. Presently, there is no specific treatment for AB except symptomatic supportive treatment. It is urgent to find an effective treatment for AB. Jinzhen Oral Liquid (JZOL) has been found to have a broad spectrum of anti-inflammatory and antiviral effects in previous clinical and basic studies and has a good effect on AB in children. However, the large-sample, randomized, double-blind, head-to-head, evidence-based studies are lacking. The purpose of this protocol is to evaluate the efficacy, safety, and mechanism of JZOL in the treatment of AB in children. Methods: This is a randomized, double-blind, parallel-controlled multi-center clinical trial. The sample size is 500 participants in the intervention group and the control group respectively, with a total of 1000 participants. They will be recruited by 10 hospitals in China. The Intervention group takes JZOL and Ambroxol Hydrochloride and Clenbuterol Hydrochloride Oral Solution (AHCHOS) placebo, while the control group receives AHCHOS and JZOL placebo. The dosage of the two drugs varies according to age and weight. The medication lasts for 7 days. The disappearance time of cough is adopted as the primary outcome. Quality control will be carried out at every stage of data management and processing to ensure that all data are reliable and processed correctly. SAS is used for statistical analysis. Intention-to-treat analysis will be carried out in this trial. All statistical tests are conducted using a two-sided test, and p <0.05 would be considered statistically significant. Discussion: We hypothesized that children with AB could get good health benefits from JZOL. This study not only evaluates the clinical efficacy and safety of JZOL but also conducts metagenomics analysis and metabolomics analysis of feces and saliva of participants to study the mechanism of JZOL against AB. Therefore, this protocol evaluates the efficacy, safety, and mechanism of JZOL from a comprehensive perspective, so as to obtain a more solid evidence chain, which will enhance the credibility of the evidence. If successful, this study will provide a high-level evidence-based reference for the treatment of AB in children and future relevant studies.
Collapse
Affiliation(s)
- Xin Cui
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Long Liang
- First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Hongjiao Geng
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junyu Xi
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Junhong Wang
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | | | - Eow Gaik Bee
- Department of Cerebral Neurology, Penang General Hospital, Penang, Malaysia
| | - Yan Chai
- Department of Epidemiology, University of California, Los Angeles, Los Angeles, United States
| | - ShengXian Wu
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: YanMing Xie, ; De Jin, ; ShengXian Wu,
| | - De Jin
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
- *Correspondence: YanMing Xie, ; De Jin, ; ShengXian Wu,
| | - YanMing Xie
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: YanMing Xie, ; De Jin, ; ShengXian Wu,
| |
Collapse
|
141
|
Nguyen PH, Le AH, Pek JSQ, Pham TT, Jayasinghe MK, Do DV, Phung CD, Le MT. Extracellular vesicles and lipoproteins - Smart messengers of blood cells in the circulation. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e49. [PMID: 38938581 PMCID: PMC11080875 DOI: 10.1002/jex2.49] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/12/2022] [Accepted: 06/19/2022] [Indexed: 06/29/2024]
Abstract
Blood cell-derived extracellular vesicles (BCEVs) and lipoproteins are the major circulating nanoparticles in blood that play an important role in intercellular communication. They have attracted significant interest for clinical applications, given their endogenous characteristics which make them stable, biocompatible, well tolerated, and capable of permeating biological barriers efficiently. In this review, we describe the basic characteristics of BCEVs and lipoproteins and summarize their implications in both physiological and pathological processes. We also outline well accepted workflows for the isolation and characterization of these circulating nanoparticles. Importantly, we highlight the latest progress and challenges associated with the use of circulating nanoparticles as diagnostic biomarkers and therapeutic interventions in multiple diseases. We spotlight novel engineering approaches and designs to facilitate the development of these nanoparticles by enhancing their stability, targeting capability, and delivery efficiency. Therefore, the present work provides a comprehensive overview of composition, biogenesis, functions, and clinical translation of circulating nanoparticles from the bench to the bedside.
Collapse
Affiliation(s)
- Phuong H.D. Nguyen
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Anh Hong Le
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Jonetta Shi Qi Pek
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Thach Tuan Pham
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Migara Kavishka Jayasinghe
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology ProgrammeCancer Programme and Nanomedicine Translational ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of SurgeryYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Dang Vinh Do
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Cao Dai Phung
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| | - Minh T.N. Le
- Department of Pharmacology and Institute for Digital MedicineYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Immunology ProgrammeCancer Programme and Nanomedicine Translational ProgrammeYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- Department of SurgeryYong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
| |
Collapse
|
142
|
Keskinidou C, Vassiliou AG, Dimopoulou I, Kotanidou A, Orfanos SE. Mechanistic Understanding of Lung Inflammation: Recent Advances and Emerging Techniques. J Inflamm Res 2022; 15:3501-3546. [PMID: 35734098 PMCID: PMC9207257 DOI: 10.2147/jir.s282695] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening lung injury characterized by an acute inflammatory response in the lung parenchyma. Hence, it is considered as the most appropriate clinical syndrome to study pathogenic mechanisms of lung inflammation. ARDS is associated with increased morbidity and mortality in the intensive care unit (ICU), while no effective pharmacological treatment exists. It is very important therefore to fully characterize the underlying pathobiology and the related mechanisms, in order to develop novel therapeutic approaches. In vivo and in vitro models are important pre-clinical tools in biological and medical research in the mechanistic and pathological understanding of the majority of diseases. In this review, we will present data from selected experimental models of lung injury/acute lung inflammation, which have been based on clinical disorders that can lead to the development of ARDS and related inflammatory lung processes in humans, including ventilation-induced lung injury (VILI), sepsis, ischemia/reperfusion, smoke, acid aspiration, radiation, transfusion-related acute lung injury (TRALI), influenza, Streptococcus (S.) pneumoniae and coronaviruses infection. Data from the corresponding clinical conditions will also be presented. The mechanisms related to lung inflammation that will be covered are oxidative stress, neutrophil extracellular traps, mitogen-activated protein kinase (MAPK) pathways, surfactant, and water and ion channels. Finally, we will present a brief overview of emerging techniques in the field of omics research that have been applied to ARDS research, encompassing genomics, transcriptomics, proteomics, and metabolomics, which may recognize factors to help stratify ICU patients at risk, predict their prognosis, and possibly, serve as more specific therapeutic targets.
Collapse
Affiliation(s)
- Chrysi Keskinidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Alice G Vassiliou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Ioanna Dimopoulou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Anastasia Kotanidou
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| | - Stylianos E Orfanos
- First Department of Critical Care Medicine and Pulmonary Services, School of Medicine, National and Kapodistrian University of Athens, "Evangelismos" Hospital, Athens, Greece
| |
Collapse
|
143
|
Simicic D, Cudalbu C, Pierzchala K. Overview of oxidative stress findings in hepatic encephalopathy: From cellular and ammonium-based animal models to human data. Anal Biochem 2022; 654:114795. [PMID: 35753389 DOI: 10.1016/j.ab.2022.114795] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 05/26/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022]
Abstract
Oxidative stress is a natural phenomenon in the body. Under physiological conditions intracellular reactive oxygen species (ROS) are normal components of signal transduction cascades, and their levels are maintained by a complex antioxidants systems participating in the in-vivo redox homeostasis. Increased oxidative stress is present in several chronic diseases and interferes with phagocytic and nervous cell functions, causing an up-regulation of cytokines and inflammation. Hepatic encephalopathy (HE) occurs in both acute liver failure (ALF) and chronic liver disease. Increased blood and brain ammonium has been considered as an important factor in pathogenesis of HE and has been associated with inflammation, neurotoxicity, and oxidative stress. The relationship between ROS and the pathophysiology of HE is still poorly understood. Therefore, sensing ROS production for a better understanding of the relationship between oxidative stress and functional outcome in HE pathophysiology is critical for determining the disease mechanisms, as well as to improve the management of patients. This review is emphasizing the important role of oxidative stress in HE development and documents the changes occurring as a consequence of oxidative stress augmentation based on cellular and ammonium-based animal models to human data.
Collapse
Affiliation(s)
- D Simicic
- CIBM Center for Biomedical Imaging, Switzerland; Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Laboratory of Functional and Metabolic Imaging, EPFL, Lausanne, Switzerland
| | - C Cudalbu
- CIBM Center for Biomedical Imaging, Switzerland; Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - K Pierzchala
- CIBM Center for Biomedical Imaging, Switzerland; Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland; Laboratory of Functional and Metabolic Imaging, EPFL, Lausanne, Switzerland.
| |
Collapse
|
144
|
Downey J, Randolph HE, Pernet E, Tran KA, Khader SA, King IL, Barreiro LB, Divangahi M. Mitochondrial cyclophilin D promotes disease tolerance by licensing NK cell development and IL-22 production against influenza virus. Cell Rep 2022; 39:110974. [PMID: 35732121 DOI: 10.1016/j.celrep.2022.110974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/25/2022] [Accepted: 05/26/2022] [Indexed: 11/03/2022] Open
Abstract
Severity of pulmonary viral infections, including influenza A virus (IAV), is linked to excessive immunopathology, which impairs lung function. Thus, the same immune responses that limit viral replication can concomitantly cause lung damage that must be countered by largely uncharacterized disease tolerance mechanisms. Here, we show that mitochondrial cyclophilin D (CypD) protects against IAV via disease tolerance. CypD-/- mice are significantly more susceptible to IAV infection despite comparable antiviral immunity. This susceptibility results from damage to the lung epithelial barrier caused by a reduction in interleukin-22 (IL-22)-producing natural killer (NK) cells. Transcriptomic and functional data reveal that CypD-/- NK cells are immature and have altered cellular metabolism and impaired IL-22 production, correlating with dysregulated bone marrow lymphopoiesis. Administration of recombinant IL-22 or transfer of wild-type (WT) NK cells abrogates pulmonary damage and protects CypD-/- mice after IAV infection. Collectively, these results demonstrate a key role for CypD in NK cell-mediated disease tolerance.
Collapse
Affiliation(s)
- Jeffrey Downey
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Haley E Randolph
- Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Erwan Pernet
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Kim A Tran
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Shabaana A Khader
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Irah L King
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada
| | - Luis B Barreiro
- Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA; Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Maziar Divangahi
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada; Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, 1001 Decarie Boulevard, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
145
|
Yao Y, Liu H, Yuan L, Du X, Yang Y, Zhou K, Wu X, Qin L, Yang M, Xiang Y, Qu X, Qin X, Liu C. Integrins are double-edged swords in pulmonary infectious diseases. Biomed Pharmacother 2022; 153:113300. [PMID: 35728353 DOI: 10.1016/j.biopha.2022.113300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Integrins are an important family of adhesion molecules that are widely distributed on immune cells in the lungs. Of note, accumulating evidences have shown that integrins are double-edged swords in pulmonary infectious diseases. On one hand, integrins promote the migration of immune cells to remove the invaded pathogens in the infected lungs. However, on the other hand, integrins also act as the targets for pathogens to escape from host immune system, which is a potential factor leading to further tissue damage. Thus, the innovative therapeutic strategies based on integrins has inspired well-founded hopes to treat pulmonary infectious diseases. In this review, we illustrate the involvement of integrins in pulmonary infectious diseases, and further discuss the innovative therapeutic targets based on integrins.
Collapse
Affiliation(s)
- Ye Yao
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Yu Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Kai Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xinyu Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Ling Qin
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China
| | - Chi Liu
- Department of Respiratory Medicine, National Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, China; Research Center of China-Africa Infectious Diseases, Xiangya School of Medicine Central South University, Changsha, Hunan, China.
| |
Collapse
|
146
|
Stevens J, Steinmeyer S, Bonfield M, Peterson L, Wang T, Gray J, Lewkowich I, Xu Y, Du Y, Guo M, Wynn JL, Zacharias W, Salomonis N, Miller L, Chougnet C, O’Connor DH, Deshmukh H. The balance between protective and pathogenic immune responses to pneumonia in the neonatal lung is enforced by gut microbiota. Sci Transl Med 2022; 14:eabl3981. [PMID: 35704600 PMCID: PMC10032669 DOI: 10.1126/scitranslmed.abl3981] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Although modern clinical practices such as cesarean sections and perinatal antibiotics have improved infant survival, treatment with broad-spectrum antibiotics alters intestinal microbiota and causes dysbiosis. Infants exposed to perinatal antibiotics have an increased likelihood of life-threatening infections, including pneumonia. Here, we investigated how the gut microbiota sculpt pulmonary immune responses, promoting recovery and resolution of infection in newborn rhesus macaques. Early-life antibiotic exposure interrupted the maturation of intestinal commensal bacteria and disrupted the developmental trajectory of the pulmonary immune system, as assessed by single-cell proteomic and transcriptomic analyses. Early-life antibiotic exposure rendered newborn macaques more susceptible to bacterial pneumonia, concurrent with increases in neutrophil senescence and hyperinflammation, broad inflammatory cytokine signaling, and macrophage dysfunction. This pathogenic reprogramming of pulmonary immunity was further reflected by a hyperinflammatory signature in all pulmonary immune cell subsets coupled with a global loss of tissue-protective, homeostatic pathways in the lungs of dysbiotic newborns. Fecal microbiota transfer was associated with partial correction of the broad immune maladaptations and protection against severe pneumonia. These data demonstrate the importance of intestinal microbiota in programming pulmonary immunity and support the idea that gut microbiota promote the balance between pathways driving tissue repair and inflammatory responses associated with clinical recovery from infection in infants. Our results highlight a potential role for microbial transfer for immune support in these at-risk infants.
Collapse
Affiliation(s)
- Joseph Stevens
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Medical Scientist Training Program, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Shelby Steinmeyer
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Madeline Bonfield
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Laura Peterson
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Timothy Wang
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jerilyn Gray
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Ian Lewkowich
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yan Xu
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Bioinformatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yina Du
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Minzhe Guo
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - James L. Wynn
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - William Zacharias
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Bioinformatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lisa Miller
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA 95616, USA
- California National Primate Research Center, Davis, CA 95616, USA
| | - Claire Chougnet
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Dennis Hartigan O’Connor
- California National Primate Research Center, Davis, CA 95616, USA
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA 95616, USA
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Hitesh Deshmukh
- Division of Neonatology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Corresponding author.
| |
Collapse
|
147
|
Rubio-Casillas A, Gupta RC, Redwa EM, Uversky VN, Badierah R. Early taurine administration as a means for halting the cytokine storm progression in COVID-19 patients. EXPLORATION OF MEDICINE 2022:234-248. [DOI: 10.37349/emed.2022.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 05/09/2022] [Indexed: 01/04/2025] Open
Abstract
Around the world, more than 6.2 million individuals have died as a result of coronavirus disease 2019 (COVID-19). According to a recent survey conducted among immunologists, epidemiologists, and virologists, this disease is expected to become endemic. This implies that the disease could have a continuous presence and/or normal frequency in the population. Pharmacological interventions to prevent infection, as well as to treat the patients at an early phase of illness to avoid hospitalization are essential additions to the vaccines. Taurine is known to inhibit the generation of all inflammatory mediators linked to the cytokine storm. It can also protect against lung injury by suppressing increased oxidants production and promoting the resolution of the inflammatory process. Neutrophil lactoferrin degranulation stimulated by taurine may have antiviral effects against SARS-CoV-2, limiting viral replication. It is hypothesized that if taurine is administered early in the onset of COVID-19 disease, it may stop the cytokine storm from progressing, lowering morbidity and mortality.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- 1Autlán Regional Hospital, Health Secretariat, Autlán, Jalisco 48900, Mexico 2Biology Laboratory, Autlán Regional High School, University of Guadalajara, Autlán, Jalisco 48900, Mexico
| | - Ramesh C. Gupta
- 3School of Agricultural Sciences and Rural Development, Nagaland University, Medziphema 797004, India
| | - Elrashdy M. Redwa
- 4Biological Science Department, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia 5Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria 21934, Egypt
| | - Vladimir N. Uversky
- 6Department of Molecular Medicine and USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Raied Badierah
- 7Medical Laboratory, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
148
|
Liew YJM, Ibrahim PAS, Ong HM, Chong CN, Tan CT, Schee JP, Gómez Román R, Cherian NG, Wong WF, Chang LY. The Immunobiology of Nipah Virus. Microorganisms 2022; 10:microorganisms10061162. [PMID: 35744680 PMCID: PMC9228579 DOI: 10.3390/microorganisms10061162] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/31/2022] [Accepted: 06/03/2022] [Indexed: 12/23/2022] Open
Abstract
Nipah virus (NiV) is a highly lethal zoonotic paramyxovirus that emerged in Malaysia in 1998. It is a human pathogen capable of causing severe respiratory infection and encephalitis. The natural reservoir of NiV, Pteropus fruit bats, remains a continuous virus source for future outbreaks, although infection in the bats is largely asymptomatic. NiV provokes serious disease in various mammalian species. In the recent human NiV outbreaks in Bangladesh and India, both bats-to-human and human-to-human transmissions have been observed. NiV has been demonstrated to interfere with the innate immune response via interferon type I signaling, promoting viral dissemination and preventing antiviral response. Studies of humoral immunity in infected NiV patients and animal models have shown that NiV-specific antibodies were produced upon infection and were protective. Studies on cellular immunity response to NiV infection in human and animal models also found that the adaptive immune response, specifically CD4+ and CD8+ T cells, was stimulated upon NiV infection. The experimental vaccines and therapeutic strategies developed have provided insights into the immunological requirements for the development of successful medical countermeasures against NiV. This review summarizes the current understanding of NiV pathogenesis and innate and adaptive immune responses induced upon infection.
Collapse
Affiliation(s)
- Yvonne Jing Mei Liew
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
- Deputy Vice Chancellor’s Office (Research & Innovation), Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Puteri Ainaa S. Ibrahim
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Hui Ming Ong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Chee Ning Chong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Chong Tin Tan
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (C.T.T.); (J.P.S.)
| | - Jie Ping Schee
- Division of Neurology, Department of Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (C.T.T.); (J.P.S.)
| | - Raúl Gómez Román
- Vaccine Research and Development, Coalition for Epidemic Preparedness Innovation (CEPI), Askekroken 11, 0277 Oslo, Norway; (R.G.R.); (N.G.C.)
| | - Neil George Cherian
- Vaccine Research and Development, Coalition for Epidemic Preparedness Innovation (CEPI), Askekroken 11, 0277 Oslo, Norway; (R.G.R.); (N.G.C.)
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
| | - Li-Yen Chang
- Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia; (Y.J.M.L.); (P.A.S.I.); (H.M.O.); (C.N.C.); (W.F.W.)
- Correspondence:
| |
Collapse
|
149
|
Sabbatinelli J, Matacchione G, Giuliani A, Ramini D, Rippo MR, Procopio AD, Bonafè M, Olivieri F. Circulating biomarkers of inflammaging as potential predictors of COVID-19 severe outcomes. Mech Ageing Dev 2022; 204:111667. [PMID: 35341896 PMCID: PMC8949647 DOI: 10.1016/j.mad.2022.111667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/22/2022] [Accepted: 03/22/2022] [Indexed: 01/10/2023]
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 infection has been of unprecedented clinical and socio-economic worldwide relevance. The case fatality rate for COVID-19 grows exponentially with age and the presence of comorbidities. In the older patients, COVID-19 manifests predominantly as a systemic disease associated with immunological, inflammatory, and procoagulant responses. Timely diagnosis and risk stratification are crucial steps to define appropriate therapies and reduce mortality, especially in the older patients. Chronically and systemically activated innate immune responses and impaired antiviral responses have been recognized as the results of a progressive remodeling of the immune system during aging, which can be described by the words 'immunosenescence' and 'inflammaging'. These age-related features of the immune system were highlighted in patients affected by COVID-19 with the poorest clinical outcomes, suggesting that the mechanisms underpinning immunosenescence and inflammaging could be relevant for COVID-19 pathogenesis and progression. Increasing evidence suggests that senescent myeloid and endothelial cells are characterized by the acquisition of a senescence-associated pro-inflammatory phenotype (SASP), which is considered as the main culprit of both immunosenescence and inflammaging. Here, we reviewed this evidence and highlighted several circulating biomarkers of inflammaging that could provide additional prognostic information to stratify COVID-19 patients based on the risk of severe outcomes.
Collapse
Affiliation(s)
- Jacopo Sabbatinelli
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Laboratory Medicine, AOU Ospedali Riuniti, Ancona, Italy
| | - Giulia Matacchione
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Deborah Ramini
- Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Maria Rita Rippo
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy
| | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine, Università di Bologna, Bologna, Italy
| | - Fabiola Olivieri
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy; Center of Clinical Pathology and Innovative Therapy, IRCCS INRCA, Ancona, Italy.
| |
Collapse
|
150
|
Zhang Y, Wang Q, Mackay CR, Ng LG, Kwok I. Neutrophil subsets and their differential roles in viral respiratory diseases. J Leukoc Biol 2022; 111:1159-1173. [PMID: 35040189 PMCID: PMC9015493 DOI: 10.1002/jlb.1mr1221-345r] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 12/19/2022] Open
Abstract
Neutrophils play significant roles in immune homeostasis and as neutralizers of microbial infections. Recent evidence further suggests heterogeneity of neutrophil developmental and activation states that exert specialized effector functions during inflammatory disease conditions. Neutrophils can play multiple roles during viral infections, secreting inflammatory mediators and cytokines that contribute significantly to host defense and pathogenicity. However, their roles in viral immunity are not well understood. In this review, we present an overview of neutrophil heterogeneity and its impact on the course and severity of viral respiratory infectious diseases. We focus on the evidence demonstrating the crucial roles neutrophils play in the immune response toward respiratory infections, using influenza as a model. We further extend the understanding of neutrophil function with the studies pertaining to COVID-19 disease and its neutrophil-associated pathologies. Finally, we discuss the relevance of these results for future therapeutic options through targeting and regulating neutrophil-specific responses.
Collapse
Affiliation(s)
- Yuning Zhang
- Department of ResearchNational Skin CentreSingaporeSingapore
| | - Quanbo Wang
- School of Pharmaceutical Sciences, Shandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Charles R Mackay
- School of Pharmaceutical Sciences, Shandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
- Department of Microbiology, Infection and Immunity ProgramBiomedicine Discovery Institute, Monash UniversityMelbourneAustralia
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN)A*STAR (Agency for Science, Technology and Research)BiopolisSingapore
- State Key Laboratory of Experimental HematologyInstitute of Hematology, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Department of Microbiology and ImmunologyImmunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of SingaporeSingaporeSingapore
- National Cancer Centre SingaporeSingaporeSingapore
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN)A*STAR (Agency for Science, Technology and Research)BiopolisSingapore
| |
Collapse
|