51
|
Heikema AP, Strepis N, Horst-Kreft D, Huynh S, Zomer A, Kelly DJ, Cooper KK, Parker CT. Biomolecule sulphation and novel methylations related to Guillain-Barré syndrome-associated Campylobacter jejuni serotype HS:19. Microb Genom 2021; 7. [PMID: 34723785 PMCID: PMC8743553 DOI: 10.1099/mgen.0.000660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Campylobacter jejuni strains that produce sialylated lipooligosaccharides (LOS) can cause the immune-mediated disease Guillain-Barré syndrome (GBS). The risk of GBS after infection with C. jejuni Penner serotype HS:19 is estimated to be at least six times higher than the average risk. Aside from LOS biosynthesis genes, genomic characteristics that promote an increased risk for GBS following C. jejuni HS:19 infection, remain uncharacterized. We hypothesized that strains with the HS:19 serotype have unique genomic features that explain the increased risk for GBS. We performed genome sequencing, alignments, single nucleotide polymorphisms' analysis and methylome characterization on a subset, and pan-genome analysis on a large number of genomes to compare HS:19 with non-HS:19 C. jejuni genome sequences. Comparison of 36 C. jejuni HS:19 with 874 C. jejuni non-HS:19 genome sequences led to the identification of three single genes and ten clusters containing contiguous genes that were significantly associated with C. jejuni HS:19. One gene cluster of seven genes, localized downstream of the capsular biosynthesis locus, was related to sulphation of biomolecules. This cluster also encoded the campylobacter sialyl transferase Cst-I. Interestingly, sulphated bacterial biomolecules such as polysaccharides can promote immune responses and, therefore, (in the presence of sialic acid) may play a role in the development of GBS. Additional gene clusters included those involved in persistence-mediated pathogenicity and gene clusters involved in restriction-modification systems. Furthermore, characterization of methylomes of two HS:19 strains exhibited novel methylation patterns (5′-CATG-3 and 5′-m6AGTNNNNNNRTTG-3) that could differentially effect gene-expression patterns of C. jejuni HS:19 strains. Our study provides novel insight into specific genetic features and possible virulence factors of C. jejuni associated with the HS:19 serotype that may explain the increased risk of GBS.
Collapse
Affiliation(s)
- Astrid P. Heikema
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), Rotterdam, The Netherlands
- *Correspondence: Astrid P. Heikema,
| | - Nikolaos Strepis
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), Rotterdam, The Netherlands
| | - Deborah Horst-Kreft
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre (Erasmus MC), Rotterdam, The Netherlands
| | - Steven Huynh
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, California, USA
| | - Aldert Zomer
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - David J. Kelly
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
| | - Kerry K. Cooper
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona, USA
| | - Craig T. Parker
- Produce Safety and Microbiology Research Unit, Agricultural Research Service, United States Department of Agriculture, Albany, California, USA
- *Correspondence: Craig T. Parker,
| |
Collapse
|
52
|
Sollberger G. Approaching Neutrophil Pyroptosis. J Mol Biol 2021; 434:167335. [PMID: 34757055 DOI: 10.1016/j.jmb.2021.167335] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 01/21/2023]
Abstract
All cells must die at some point, and the dogma is that they do it either silently via apoptosis or via pro-inflammatory, lytic forms of death. Amongst these lytic cell death pathways, pyroptosis is one of the best characterized. Pyroptosis depends on inflammatory caspases which activate members of the gasdermin family of proteins, and it is associated with the release of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18. Pyroptosis is an essential component of innate immunity, it initiates and amplifies inflammation and it removes the replication niche for intracellular pathogens. Most of the literature on pyroptosis focuses on monocytes and macrophages. However, the most abundant phagocytes in humans are neutrophils. This review addresses whether neutrophils undergo pyroptosis and the underlying mechanisms. Furthermore, I discuss how and why neutrophils might be able to resist pyroptosis.
Collapse
Affiliation(s)
- Gabriel Sollberger
- University of Dundee, School of Life Sciences, Division of Cell Signalling and Immunology, Dow Street, DD1 5EH Dundee, UK; Max Planck Institute for Infection Biology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
53
|
Karnaushkina MA, Guryev AS, Mironov KO, Dunaeva EA, Korchagin VI, Bobkova OY, Vasilyeva IS, Kassina DV, Litvinova MM. Associations of Toll-like Receptor Gene Polymorphisms with NETosis Activity as Prognostic Criteria for the Severity of Pneumonia. Sovrem Tekhnologii Med 2021; 13:47-53. [PMID: 34603755 PMCID: PMC8482823 DOI: 10.17691/stm2021.13.3.06] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Indexed: 12/02/2022] Open
Abstract
The aim of the study was to determine the molecular genetic prognostic criteria for the severity of the course pneumonia based on the analysis of the association of genetic polymorphism in toll-like receptors with the severity of NETosis.
Collapse
Affiliation(s)
- M A Karnaushkina
- Professor, Department of Internal Diseases with a Course of Cardiology and Functional Diagnostics named after Academician V.S. Moiseev; Peoples' Friendship University of Russia, 6 Miklukho-Maklaya St., Moscow, 117198, Russia
| | - A S Guryev
- Senior Researcher, Research Laboratory; Moscow Regional Research Clinical Institute named after M.F. Vladimirsky, 61/2-1 Schepkina St., Moscow, 129110, Russia
| | - K O Mironov
- Head of the Research Group for the Development of New Methods for Identifying Genetic Polymorphisms; Central Research Institute of Epidemiology of the Federal Service on Consumer Rights Protection and Human Wellbeing (Rospotrebnadzor), 3а Novogireevskaya St., Moscow, 111123, Russia
| | - E A Dunaeva
- Researcher, Research Group for the Development of New Methods for Identifying Genetic Polymorphisms; Central Research Institute of Epidemiology of the Federal Service on Consumer Rights Protection and Human Wellbeing (Rospotrebnadzor), 3а Novogireevskaya St., Moscow, 111123, Russia
| | - V I Korchagin
- Researcher, Research Group for the Development of New Methods for Identifying Genetic Polymorphisms; Central Research Institute of Epidemiology of the Federal Service on Consumer Rights Protection and Human Wellbeing (Rospotrebnadzor), 3а Novogireevskaya St., Moscow, 111123, Russia
| | - O Yu Bobkova
- PhD Student, Department of Hospital Therapy No.2; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya St., Moscow, 119991, Russia
| | - I S Vasilyeva
- Assistant, Department of Hospital Therapy No.2; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya St., Moscow, 119991, Russia
| | - D V Kassina
- Researcher, Research Laboratory; Moscow Regional Research Clinical Institute named after M.F. Vladimirsky, 61/2-1 Schepkina St., Moscow, 129110, Russia
| | - M M Litvinova
- Associate Professor, Department of Medical Genetics; I.M. Sechenov First Moscow State Medical University (Sechenov University), 8/2 Trubetskaya St., Moscow, 119991, Russia; Geneticist, Center for Personalized Medicine; Moscow Clinical Scientific Center named after A.S. Loginov, Moscow Healthcare Department, 86 Shosse Entuziastov, Moscow, 111123, Russia
| |
Collapse
|
54
|
|
55
|
Monteith AJ, Miller JM, Maxwell CN, Chazin WJ, Skaar EP. Neutrophil extracellular traps enhance macrophage killing of bacterial pathogens. SCIENCE ADVANCES 2021; 7:eabj2101. [PMID: 34516771 PMCID: PMC8442908 DOI: 10.1126/sciadv.abj2101] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/20/2021] [Indexed: 05/16/2023]
Abstract
Neutrophils and macrophages are critical to the innate immune response, but cooperative mechanisms used by these cells to combat extracellular pathogens are not well understood. This study reveals that S100A9-deficient neutrophils produce higher levels of mitochondrial superoxide in response to Staphylococcus aureus and, as a result, form neutrophil extracellular traps (suicidal NETosis). Increased suicidal NETosis does not improve neutrophil killing of S. aureus in isolation but augments macrophage killing. NET formation enhances antibacterial activity by increasing phagocytosis by macrophages and by transferring neutrophil-specific antimicrobial peptides to them. Similar results were observed in response to other phylogenetically distinct bacterial pathogens including Streptococcus pneumoniae and Pseudomonas aeruginosa, implicating this as an immune defense mechanism that broadly enhances antibacterial activity. These results demonstrate that achieving maximal bactericidal activity through NET formation requires macrophages and that accelerated and more robust suicidal NETosis makes neutrophils adept at increasing antibacterial activity, especially when A9 deficient.
Collapse
Affiliation(s)
- Andrew J. Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeanette M. Miller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - C. Noel Maxwell
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Walter J. Chazin
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
56
|
Agak GW, Mouton A, Teles RM, Weston T, Morselli M, Andrade PR, Pellegrini M, Modlin RL. Extracellular traps released by antimicrobial TH17 cells contribute to host defense. J Clin Invest 2021; 131:141594. [PMID: 33211671 DOI: 10.1172/jci141594] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/12/2020] [Indexed: 12/19/2022] Open
Abstract
TH17 cell subpopulations have been defined that contribute to inflammation and homeostasis, yet the characteristics of TH17 cells that contribute to host defense against infection are not clear. To elucidate the antimicrobial machinery of the TH17 subset, we studied the response to Cutibacterium acnes, a skin commensal that is resistant to IL-26, the only known TH17-secreted protein with direct antimicrobial activity. We generated C. acnes-specific antimicrobial TH17 clones (AMTH17) with varying antimicrobial activity against C. acnes, which we correlated by RNA sequencing to the expression of transcripts encoding proteins that contribute to antimicrobial activity. Additionally, we validated that AMTH17-mediated killing of C. acnes and bacterial pathogens was dependent on the secretion of granulysin, granzyme B, perforin, and histone H2B. We found that AMTH17 cells can release fibrous structures composed of DNA decorated with histone H2B that entangle C. acnes that we call T cell extracellular traps (TETs). Within acne lesions, H2B and IL-17 colocalized in CD4+ T cells, in proximity to TETs in the extracellular space composed of DNA decorated with H2B. This study identifies a functionally distinct subpopulation of TH17 cells with an ability to form TETs containing secreted antimicrobial proteins that capture and kill bacteria.
Collapse
Affiliation(s)
- George W Agak
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Alice Mouton
- Department of Ecology and Evolutionary Biology, UCLA, Los Angeles, California, USA
| | - Rosane Mb Teles
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Thomas Weston
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, and.,Institute for Quantitative and Computational Biosciences - The Collaboratory, UCLA, Los Angeles, California, USA
| | - Priscila R Andrade
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, and.,Institute for Quantitative and Computational Biosciences - The Collaboratory, UCLA, Los Angeles, California, USA
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA.,Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| |
Collapse
|
57
|
Characteristics and Role of Neutrophil Extracellular Traps in Asthma. Inflammation 2021; 45:6-13. [PMID: 34480251 PMCID: PMC8803764 DOI: 10.1007/s10753-021-01526-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 07/22/2021] [Indexed: 12/12/2022]
Abstract
Asthma is a common chronic respiratory disease that affects millions of people worldwide. The incidence of asthma has continued to increase every year. Bronchial asthma involves a variety of cells, including airway inflammatory cells, structural cells, and neutrophils, which have gained more attention because they secrete substances that play an important role in the occurrence and development of asthma. Neutrophil extracellular traps (NETs) are mesh-like structures composed of DNA, histones, and non-histone molecules that can be secreted from neutrophils. NETs can enrich anti-bacterial substances and limit pathogen migration, thus having a protective effect in case of inflammation. However, despite of their anti-inflammatory properties, NETs have been shown to trigger allergic asthma and worsen asthma progression. Here, we provide a systematic review of the roles of NETs in asthma.
Collapse
|
58
|
Zhou X, Wang H, Lian S, Wang J, Wu R. Effect of Copper, Zinc, and Selenium on the Formation of Bovine Neutrophil Extracellular Traps. Biol Trace Elem Res 2021; 199:3312-3318. [PMID: 33180264 DOI: 10.1007/s12011-020-02477-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/03/2020] [Indexed: 10/23/2022]
Abstract
Dairy cow neutrophils activate a program leading to cell death and expulsion of neutrophil extracellular traps (NETs). The role of NETs is to capture pathogens, degrade bacterial toxic factors, and kill bacteria, and the effect of trace elements on NETs formation in cows is ambiguous. In this study, we investigated the effect of copper (0.5 mg/L, 0.8 mg/L, and 2.0 mg/L), zinc (0.1 mg/L, 1.0 mg/L, and 2.0 mg/L), and selenium (0.01 mg/L, 0.08 mg/L, and 2.0 mg/L) on NETs formation in dairy cows. Trace element induction of NETs formation was observed by laser confocal microscopy. The percentage of NETs formed was calculated by quantifying the number of neutrophils forming NETs out of the total number of neutrophils observed under 20 high-power (200×) magnification fields. Copper, zinc, and selenium induced the formation of a network of DNA, neutrophil elastase (ELA2), and myeloperoxidase. Copper (0.8 mg/L), zinc (1.0 mg/L), and selenium (0.01 mg/L) significantly induced the formation of NETs (p < 0.05). The study provides an experimental basis for enhancing the immunity of cows before and after delivery by adding copper, zinc, and selenium.
Collapse
Affiliation(s)
- Xiechen Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, Heilongjiang, People's Republic of China
| | - Hai Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, Heilongjiang, People's Republic of China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, Heilongjiang, People's Republic of China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, People's Republic of China
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, Heilongjiang, People's Republic of China
| | - Rui Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang, People's Republic of China.
- Heilongjiang Provincial Key Laboratory of Prevention and Control of Bovine Diseases, Daqing, 163319, Heilongjiang, People's Republic of China.
| |
Collapse
|
59
|
A Fragile Balance: Does Neutrophil Extracellular Trap Formation Drive Pulmonary Disease Progression? Cells 2021; 10:cells10081932. [PMID: 34440701 PMCID: PMC8394734 DOI: 10.3390/cells10081932] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils act as the first line of defense during infection and inflammation. Once activated, they are able to fulfil numerous tasks to fight inflammatory insults while keeping a balanced immune response. Besides well-known functions, such as phagocytosis and degranulation, neutrophils are also able to release "neutrophil extracellular traps" (NETs). In response to most stimuli, the neutrophils release decondensed chromatin in a NADPH oxidase-dependent manner decorated with histones and granule proteins, such as neutrophil elastase, myeloperoxidase, and cathelicidins. Although primarily supposed to prevent microbial dissemination and fight infections, there is increasing evidence that an overwhelming NET response correlates with poor outcome in many diseases. Lung-related diseases especially, such as bacterial pneumonia, cystic fibrosis, chronic obstructive pulmonary disease, aspergillosis, influenza, and COVID-19, are often affected by massive NET formation. Highly vascularized areas as in the lung are susceptible to immunothrombotic events promoted by chromatin fibers. Keeping this fragile equilibrium seems to be the key for an appropriate immune response. Therapies targeting dysregulated NET formation might positively influence many disease progressions. This review highlights recent findings on the pathophysiological influence of NET formation in different bacterial, viral, and non-infectious lung diseases and summarizes medical treatment strategies.
Collapse
|
60
|
Zhang K, Jiang N, Sang X, Feng Y, Chen R, Chen Q. Trypanosoma brucei Lipophosphoglycan Induces the Formation of Neutrophil Extracellular Traps and Reactive Oxygen Species Burst via Toll-Like Receptor 2, Toll-Like Receptor 4, and c-Jun N-Terminal Kinase Activation. Front Microbiol 2021; 12:713531. [PMID: 34394064 PMCID: PMC8355521 DOI: 10.3389/fmicb.2021.713531] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/06/2021] [Indexed: 01/21/2023] Open
Abstract
Trypanosoma brucei brucei is the causative agent of African animal trypanosomosis, which mainly parasitizes the blood of the host. Lipophosphoglycan (LPG), a polymer anchored to the surface of the parasites, activates the host immune response. In this study, we revealed that T. brucei LPG stimulated neutrophils to form neutrophil extracellular traps (NETs) and release the reactive oxygen species (ROS). We further analyzed the involvement of toll-like receptor 2 (TLR2) and toll-like receptor 4 (TLR4) and explored the activation of signaling pathway enzymes in response to LPG stimulation. During the stimulation of neutrophils by LPG, the blockade using anti-TLR2 and anti-TLR4 antibodies reduced the phosphorylation of c-Jun N-terminal kinase (JNK), the release of DNA from the NETs, and the burst of ROS. Moreover, the addition of JNK inhibitor and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase inhibitor exhibited similar effects. Our data suggest that T. brucei LPG activates the phosphorylation of JNK through TLR2 and TLR4 recognition, which causes the formation of NETs and the burst of ROS.
Collapse
Affiliation(s)
- Kai Zhang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Key Laboratory of Zoonosis, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, China
| |
Collapse
|
61
|
A Review of the Neutrophil Extracellular Traps (NETs) from Cow, Sheep and Goat Models. Int J Mol Sci 2021; 22:ijms22158046. [PMID: 34360812 PMCID: PMC8347029 DOI: 10.3390/ijms22158046] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
This review provides insight into the importance of understanding NETosis in cows, sheep, and goats in light of the importance to their health, welfare and use as animal models. Neutrophils are essential to innate immunity, pathogen infection, and inflammatory diseases. The relevance of NETosis as a conserved innate immune response mechanism and the translational implications for public health are presented. Increased understanding of NETosis in ruminants will contribute to the prediction of pathologies and design of strategic interventions targeting NETs. This will help to control pathogens such as coronaviruses and inflammatory diseases such as mastitis that impact all mammals, including humans. Definition of unique attributes of NETosis in ruminants, in comparison to what has been observed in humans, has significant translational implications for one health and global food security, and thus warrants further study.
Collapse
|
62
|
The Role of Neutrophil Extracellular Traps in Central Nervous System Diseases and Prospects for Clinical Application. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9931742. [PMID: 34336122 PMCID: PMC8294981 DOI: 10.1155/2021/9931742] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 07/01/2021] [Indexed: 12/13/2022]
Abstract
Neutrophil extracellular traps (NETs) are complexes of decondensed DNA fibers and antimicrobial peptides that are released by neutrophils and play important roles in many noninfectious diseases, such as cystic fibrosis, systemic lupus erythematosus, diabetes, and cancer. Recently, the formation of NETs has been detected in many central nervous system diseases and is thought to play different roles in the occurrence and development of these diseases. Researchers have detected NETs in acute ischemic stroke thrombi, and these NETs are thought to promote coagulation and thrombosis. NETs in ischemic brain parenchyma were identified as the cause of secondary nerve damage. High levels of NETs were also detected in grade IV glioma tissues, where NETs were involved in the proliferation and invasion of glioma cells by activating a signaling pathway. Extracellular web-like structures have also recently been observed in mice with traumatic brain injury (TBI), and it was hypothesized that NETs contribute to the development of edema after TBI. This article reviews the effect of NETs on multiple diseases that affect the CNS and explores their clinical application prospects.
Collapse
|
63
|
Hirschmann S, Gómez-Mejia A, Kohler TP, Voß F, Rohde M, Brendel M, Hammerschmidt S. The Two-Component System 09 of Streptococcus pneumoniae Is Important for Metabolic Fitness and Resistance during Dissemination in the Host. Microorganisms 2021; 9:microorganisms9071365. [PMID: 34201716 PMCID: PMC8306541 DOI: 10.3390/microorganisms9071365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/12/2021] [Accepted: 06/18/2021] [Indexed: 11/24/2022] Open
Abstract
The two-component regulatory system 09 of Streptococcus pneumoniae has been shown to modulate resistance against oxidative stress as well as capsule expression. These data and the implication of TCS09 in cell wall integrity have been shown for serotype 2 strain D39. Other data have suggested strain-specific regulatory effects of TCS09. Contradictory data are known on the impact of TCS09 on virulence, but all have been explored using only the rr09-mutant. In this study, we have therefore deleted one or both components of the TCS09 (SP_0661 and SP_0662) in serotype 4 S. pneumoniae TIGR4. In vitro growth assays in chemically defined medium (CDM) using sucrose or lactose as a carbon source indicated a delayed growth of nonencapsulated tcs09-mutants, while encapsulated wild-type TIGR4 and tcs09-mutants have reduced growth in CDM with glucose. Using a set of antigen-specific antibodies, immunoblot analysis showed that only the pilus 1 backbone protein RrgB is significantly reduced in TIGR4ΔcpsΔhk09. Electron microscopy, adherence and phagocytosis assays showed no impact of TCS09 on the TIGR4 cell morphology and interaction with host cells. In contrast, in vivo infections and in particular competitive co-infection experiments demonstrated that TCS09 enhances robustness during dissemination in the host by maintaining bacterial fitness.
Collapse
Affiliation(s)
- Stephanie Hirschmann
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (T.P.K.); (F.V.); (M.B.)
| | - Alejandro Gómez-Mejia
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (T.P.K.); (F.V.); (M.B.)
| | - Thomas P. Kohler
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (T.P.K.); (F.V.); (M.B.)
| | - Franziska Voß
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (T.P.K.); (F.V.); (M.B.)
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| | - Max Brendel
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (T.P.K.); (F.V.); (M.B.)
| | - Sven Hammerschmidt
- Center for Functional Genomics of Microbes, Department of Molecular Genetics and Infection Biology, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, 17487 Greifswald, Germany; (S.H.); (A.G.-M.); (T.P.K.); (F.V.); (M.B.)
- Correspondence:
| |
Collapse
|
64
|
To Trap a Pathogen: Neutrophil Extracellular Traps and Their Role in Mucosal Epithelial and Skin Diseases. Cells 2021; 10:cells10061469. [PMID: 34208037 PMCID: PMC8230648 DOI: 10.3390/cells10061469] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 12/18/2022] Open
Abstract
Neutrophils are the most abundant circulating innate immune cells and comprise the first immune defense line, as they are the most rapidly recruited cells at sites of infection or inflammation. Their main microbicidal mechanisms are degranulation, phagocytosis, cytokine secretion and the formation of extracellular traps. Neutrophil extracellular traps (NETs) are a microbicidal mechanism that involves neutrophil death. Since their discovery, in vitro and in vivo neutrophils have been challenged with a range of stimuli capable of inducing or inhibiting NET formation, with the objective to understand its function and regulation in health and disease. These networks composed of DNA and granular components are capable of immobilizing and killing pathogens. They comprise enzymes such as myeloperoxidase, elastase, cathepsin G, acid hydrolases and cationic peptides, all with antimicrobial and antifungal activity. Therefore, the excessive formation of NETs can also lead to tissue damage and promote local and systemic inflammation. Based on this concept, in this review, we focus on the role of NETs in different infectious and inflammatory diseases of the mucosal epithelia and skin.
Collapse
|
65
|
Fantone K, Tucker SL, Miller A, Yadav R, Bernardy EE, Fricker R, Stecenko AA, Goldberg JB, Rada B. Cystic Fibrosis Sputum Impairs the Ability of Neutrophils to Kill Staphylococcus aureus. Pathogens 2021; 10:pathogens10060703. [PMID: 34200034 PMCID: PMC8229215 DOI: 10.3390/pathogens10060703] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis (CF) airway disease is characterized by chronic microbial infections and infiltration of inflammatory polymorphonuclear (PMN) granulocytes. Staphylococcus aureus (S. aureus) is a major lung pathogen in CF that persists despite the presence of PMNs and has been associated with CF lung function decline. While PMNs represent the main mechanism of the immune system to kill S. aureus, it remains largely unknown why PMNs fail to eliminate S. aureus in CF. The goal of this study was to observe how the CF airway environment affects S. aureus killing by PMNs. PMNs were isolated from the blood of healthy volunteers and CF patients. Clinical isolates of S. aureus were obtained from the airways of CF patients. The results show that PMNs from healthy volunteers were able to kill all CF isolates and laboratory strains of S. aureus tested in vitro. The extent of killing varied among strains. When PMNs were pretreated with supernatants of CF sputum, S. aureus killing was significantly inhibited suggesting that the CF airway environment compromises PMN antibacterial functions. CF blood PMNs were capable of killing S. aureus. Although bacterial killing was inhibited with CF sputum, PMN binding and phagocytosis of S. aureus was not diminished. The S. aureus-induced respiratory burst and neutrophil extracellular trap release from PMNs also remained uninhibited by CF sputum. In summary, our data demonstrate that the CF airway environment limits killing of S. aureus by PMNs and provides a new in vitro experimental model to study this phenomenon and its mechanism.
Collapse
Affiliation(s)
- Kayla Fantone
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Samantha L. Tucker
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Arthur Miller
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Ruchi Yadav
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Eryn E. Bernardy
- Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (E.E.B.); (A.A.S.); (J.B.G.)
| | - Rachel Fricker
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Arlene A. Stecenko
- Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (E.E.B.); (A.A.S.); (J.B.G.)
| | - Joanna B. Goldberg
- Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (E.E.B.); (A.A.S.); (J.B.G.)
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
- Correspondence:
| |
Collapse
|
66
|
Hassan M, Baig AA, Attique SA, Abbas S, Khan F, Zahid S, Ain QU, Usman M, Simbak NB, Kamal MA, Yusof HA. Molecular docking of alpha-enolase to elucidate the promising candidates against Streptococcus pneumoniae infection. Daru 2021; 29:73-84. [PMID: 33537864 PMCID: PMC8149539 DOI: 10.1007/s40199-020-00384-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/23/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE To predict potential inhibitors of alpha-enolase to reduce plasminogen binding of Streptococcus pneumoniae (S. pneumoniae) that may lead as an orally active drug. S. pneumoniae remains dominant in causing invasive diseases. Fibrinolytic pathway is a critical factor of S. pneumoniae to invade and progression of disease in the host body. Besides the low mass on the cell surface, alpha-enolase possesses significant plasminogen binding among all exposed proteins. METHODS In-silico based drug designing approach was implemented for evaluating potential inhibitors against alpha-enolase based on their binding affinities, energy score and pharmacokinetics. Lipinski's rule of five (LRo5) and Egan's (Brain Or IntestinaL EstimateD) BOILED-Egg methods were executed to predict the best ligand for biological systems. RESULTS Molecular docking analysis revealed, Sodium (1,5-dihydroxy-2-oxopyrrolidin-3-yl)-hydroxy-dioxidophosphanium (SF-2312) as a promising inhibitor that fabricates finest attractive charges and conventional hydrogen bonds with S. pneumoniae alpha-enolase. Moreover, the pharmacokinetics of SF-2312 predict it as a therapeutic inhibitor for clinical trials. Like SF-2312, phosphono-acetohydroxamate (PhAH) also constructed adequate interactions at the active site of alpha-enolase, but it predicted less favourable than SF-2312 based on binding affinity. CONCLUSION Briefly, SF-2312 and PhAH ligands could inhibit the role of alpha-enolase to restrain plasminogen binding, invasion and progression of S. pneumoniae. As per our investigation and analysis, SF-2312 is the most potent naturally existing inhibitor of S. pneumoniae alpha-enolase in current time.
Collapse
Affiliation(s)
- Muhammad Hassan
- Faculty of Medicine, Universiti Sultan Zainal Abidin (UniSZA), Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu Darul Iman Malaysia
| | - Atif Amin Baig
- Faculty of Medicine, Universiti Sultan Zainal Abidin (UniSZA), Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu Darul Iman Malaysia
| | - Syed Awais Attique
- Research Centre for Modelling and Simulation (RCMS), National University of Sciences and Technology (NUST), Islamabad, H-12 Pakistan
| | - Shafqat Abbas
- School of Environmental Engineering, Suzhou University of Science and Technology, Suzhou New District, 215009 China
| | - Fizza Khan
- Centre of Agricultural Biochemistry and Biotechnology (CABB), University of Agriculture, Faisalabad, 38000 Pakistan
| | - Sara Zahid
- Centre of Agricultural Biochemistry and Biotechnology (CABB), University of Agriculture, Faisalabad, 38000 Pakistan
| | - Qurat Ul Ain
- Centre for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, 53700 Pakistan
| | - Muhammad Usman
- Department of Computer Science, University of Agriculture, Faisalabad, 38000 Pakistan
| | - Nordin Bin Simbak
- Faculty of Medicine, Universiti Sultan Zainal Abidin (UniSZA), Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu Darul Iman Malaysia
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah, 21589 Saudi Arabia
- Enzymoics, Novel Global Community Educational Foundation, 7 Peterlee Place, Hebersham, NSW 2770 Australia
| | - Hanani Ahmad Yusof
- Kulliyyah of Allied Health Sciences, International Islamic University Malaysia (IIUM), 25200 Kuantan, Pahang Malaysia
| |
Collapse
|
67
|
Liu Q, Yi W, Jiang S, Song J, Liang P. Neutrophil Extracellular Traps Serve as Key Effector Molecules in the Protection Against Phialophora verrucosa. Mycopathologia 2021; 186:367-375. [PMID: 34013384 PMCID: PMC8249254 DOI: 10.1007/s11046-021-00554-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/10/2021] [Indexed: 12/21/2022]
Abstract
Phialophora verrucosa (P. verrucosa) is a pathogen that can cause chromoblastomycosis and phaeohyphomycosis. Recent evidence suggests that neutrophils can produce neutrophil extracellular traps (NETs) that can protect against invasive pathogens. As such, we herein explored the in vitro functional importance of P. verrucosa-induced NET formation. By assessing the co-localization of neutrophil elastase and DNA, we were able to confirm the formation of classical NETs entrapping P. verrucosa specimens. Sytox Green was then used to stain these NETs following neutrophil infection with P. verrucosa in order to quantify the formation of these extracellular structures. NET formation was induced upon neutrophil exposure to both live, UV-inactivated, and dead P. verrucosa fungi. The ability of these NETs to kill fungal hyphae and conidia was demonstrated through MTT and pouring plate assays, respectively. Overall, our results confirmed that P. verrucosa was able to trigger the production of NETs, suggesting that these extracellular structures may represent an important innate immune effector mechanism controlling physiological responses to P. verrucosa infection, thereby aiding in pathogen control during the acute phases of infection.
Collapse
Affiliation(s)
- Qin Liu
- Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenjuan Yi
- Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Si Jiang
- Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jiquan Song
- Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Pin Liang
- Department of Dermatology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
68
|
Payne JAE, Kulkarni K, Izore T, Fulcher AJ, Peleg AY, Aguilar MI, Cryle MJ, Del Borgo MP. Staphylococcus aureus entanglement in self-assembling β-peptide nanofibres decorated with vancomycin. NANOSCALE ADVANCES 2021; 3:2607-2616. [PMID: 36134162 PMCID: PMC9419598 DOI: 10.1039/d0na01018a] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/23/2021] [Indexed: 06/16/2023]
Abstract
The increasing resistance of pathogenic microbes to antimicrobials and the shortage of antibiotic drug discovery programs threaten the clinical use of antibiotics. This threat calls for the development of new methods for control of drug-resistant microbial pathogens. We have designed, synthesised and characterised an antimicrobial material formed via the self-assembly of a population of two distinct β-peptide monomers, a lipidated tri-β-peptide (β3-peptide) and a novel β3-peptide conjugated to a glycopeptide antibiotic, vancomycin. The combination of these two building blocks resulted in fibrous assemblies with distinctive structures determined by atomic force microscopy and electron microscopy. These fibres inhibited the growth of methicillin resistant Staphylococcus aureus (MRSA) and associated directly with the bacteria, acting as a peptide nanonet with fibre nucleation sites on the bacteria observed by electron microscopy and confocal microscopy. Our results provide insights into the design of peptide based supramolecular assemblies with antibacterial activity and establish an innovative strategy to develop self-assembled antimicrobial materials for future biomedical application.
Collapse
Affiliation(s)
- Jennifer A E Payne
- Infection and Immunity Program, The Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University Clayton Victoria 3800 Australia
- EMBL Australia, Monash University Clayton Victoria 3800 Australia
- The ARC Centre of Excellence for Innovations in Peptide and Protein Science, Monash University Clayton Victoria 3800 Australia
| | - Ketav Kulkarni
- Department of Biochemistry and Molecular Biology, Monash University Clayton Victoria 3800 Australia
| | - Thierry Izore
- Infection and Immunity Program, The Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University Clayton Victoria 3800 Australia
- EMBL Australia, Monash University Clayton Victoria 3800 Australia
| | - Alex J Fulcher
- Monash Micro Imaging, Monash University Clayton Victoria 3800 Australia
| | - Anton Y Peleg
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University Clayton Victoria 3800 Australia
- Department of Infectious Diseases, The Alfred Hospital, Central Clinical School, Monash University Melbourne Victoria 3004 Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Monash University Clayton Victoria 3800 Australia
| | - Max J Cryle
- Infection and Immunity Program, The Monash Biomedicine Discovery Institute, Department of Biochemistry and Molecular Biology, Monash University Clayton Victoria 3800 Australia
- EMBL Australia, Monash University Clayton Victoria 3800 Australia
- The ARC Centre of Excellence for Innovations in Peptide and Protein Science, Monash University Clayton Victoria 3800 Australia
| | - Mark P Del Borgo
- Department of Pharmacology, Monash University Clayton Victoria 3800 Australia
| |
Collapse
|
69
|
Zivkovic S, Ayazi M, Hammel G, Ren Y. For Better or for Worse: A Look Into Neutrophils in Traumatic Spinal Cord Injury. Front Cell Neurosci 2021; 15:648076. [PMID: 33967695 PMCID: PMC8100532 DOI: 10.3389/fncel.2021.648076] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/08/2021] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are short-lived cells of the innate immune system and the first line of defense at the site of an infection and tissue injury. Pattern recognition receptors on neutrophils recognize pathogen-associated molecular patterns or danger-associated molecular patterns, which recruit them to the destined site. Neutrophils are professional phagocytes with efficient granular constituents that aid in the neutralization of pathogens. In addition to phagocytosis and degranulation, neutrophils are proficient in creating neutrophil extracellular traps (NETs) that immobilize pathogens to prevent their spread. Because of the cytotoxicity of the associated granular proteins within NETs, the microbes can be directly killed once immobilized by the NETs. The role of neutrophils in infection is well studied; however, there is less emphasis placed on the role of neutrophils in tissue injury, such as traumatic spinal cord injury. Upon the initial mechanical injury, the innate immune system is activated in response to the molecules produced by the resident cells of the injured spinal cord initiating the inflammatory cascade. This review provides an overview of the essential role of neutrophils and explores the contribution of neutrophils to the pathologic changes in the injured spinal cord.
Collapse
Affiliation(s)
- Sandra Zivkovic
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Maryam Ayazi
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Grace Hammel
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL, United States
| |
Collapse
|
70
|
Development and validation of a new scoring system for the early diagnosis of tuberculous meningitis in adults. Diagn Microbiol Infect Dis 2021; 101:115393. [PMID: 34237646 DOI: 10.1016/j.diagmicrobio.2021.115393] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 03/24/2021] [Accepted: 04/08/2021] [Indexed: 11/21/2022]
Abstract
We developed and validated a new diagnostic scoring system by simultaneously comparing 28 factors (including clinical, laboratory, and imaging) of HIV uninfected adult tuberculous meningitis (TBM) with viral meningitis (VM), bacterial meningitis (BM), and cryptococcal meningitis (CM). Predictors of TBM diagnosis obtained by logistic regression. A total of 382 patients with intracranial infection participated, and eight factors were independently associated with TBM diagnosis: symptom duration, evidence of extracranial tuberculosis, cerebrospinal fluid (CSF) leukocyte, CSF neutrophil, CSF protein, low serum sodium, meningeal enhancement, and tuberculomas. Factors are assigned according to weight, a score of ≥ 5 was suggestive of TBM with a sensitivity of 85.8% and a specificity of 87.7%, and the area under the receiver operating characteristic curve was 0.923. When applied to a prospective validation cohort, this scoring model showed robust performance. Our study suggests that the application of this score can help diagnose TBM more efficiently.
Collapse
|
71
|
Endothelial dysfunction and immunothrombosis as key pathogenic mechanisms in COVID-19. Nat Rev Immunol 2021; 21:319-329. [PMID: 33824483 PMCID: PMC8023349 DOI: 10.1038/s41577-021-00536-9] [Citation(s) in RCA: 572] [Impact Index Per Article: 190.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 02/07/2023]
Abstract
Coronavirus disease 2019 (COVID-19) is a clinical syndrome caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Patients with severe disease show hyperactivation of the immune system, which can affect multiple organs besides the lungs. Here, we propose that SARS-CoV-2 infection induces a process known as immunothrombosis, in which activated neutrophils and monocytes interact with platelets and the coagulation cascade, leading to intravascular clot formation in small and larger vessels. Microthrombotic complications may contribute to acute respiratory distress syndrome (ARDS) and other organ dysfunctions. Therapeutic strategies aimed at reducing immunothrombosis may therefore be useful. Several antithrombotic and immunomodulating drugs have been proposed as candidates to treat patients with SARS-CoV-2 infection. The growing understanding of SARS-CoV-2 infection pathogenesis and how it contributes to critical illness and its complications may help to improve risk stratification and develop targeted therapies to reduce the acute and long-term consequences of this disease. Here, the authors propose that SARS-CoV-2 induces a prothrombotic state, with dysregulated immunothrombosis in lung microvessels and endothelial injury, which drive the clinical manifestations of severe COVID-19. They discuss potential antithrombotic and immunomodulating drugs that are being considered in the treatment of patients with COVID-19.
Collapse
|
72
|
Zhang K, Jiang N, Chen H, Zhang N, Sang X, Feng Y, Chen R, Chen Q. TatD DNases of African trypanosomes confer resistance to host neutrophil extracellular traps. SCIENCE CHINA. LIFE SCIENCES 2021; 64:621-632. [PMID: 33420923 DOI: 10.1007/s11427-020-1854-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 11/19/2020] [Indexed: 01/07/2023]
Abstract
African trypanosomatid parasites escape host acquired immune responses through periodic antigenic variation of their surface coat. In this study, we describe a mechanism by which the parasites counteract innate immune responses. Two TatD DNases were identified in each of Trypanosoma evansi and Trypanosoma brucei. These DNases are bivalent metal-dependent endonucleases localized in the cytoplasm and flagella of the parasites that can also be secreted by the parasites. These enzymes possess conserved functional domains and have efficient DNA hydrolysis activity. Host neutrophil extracellular traps (NETs) induced by the parasites could be hydrolyzed by native and recombinant TatD DNases. NET disruption was prevented, and the survival rate of parasites was decreased, in the presence of the DNase inhibitor aurintricarboxylic acid. These data suggest that trypanosomes can counteract host innate immune responses by active secretion of TatD DNases to degrade NETs.
Collapse
Affiliation(s)
- Kai Zhang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Hongyu Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Naiwen Zhang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
- The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China.
| |
Collapse
|
73
|
Lannes-Costa PS, de Oliveira JSS, da Silva Santos G, Nagao PE. A current review of pathogenicity determinants of Streptococcus sp. J Appl Microbiol 2021; 131:1600-1620. [PMID: 33772968 DOI: 10.1111/jam.15090] [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: 12/21/2020] [Revised: 03/16/2021] [Accepted: 03/24/2021] [Indexed: 12/16/2022]
Abstract
The genus Streptococcus comprises important pathogens, many of them are part of the human or animal microbiota. Advances in molecular genetics, taxonomic approaches and phylogenomic studies have led to the establishment of at least 100 species that have a severe impact on human health and are responsible for substantial economic losses to agriculture. The infectivity of the pathogens is linked to cell-surface components and/or secreted virulence factors. Bacteria have evolved sophisticated and multifaceted adaptation strategies to the host environment, including biofilm formation, survival within professional phagocytes, escape the host immune response, amongst others. This review focuses on virulence mechanism and zoonotic potential of Streptococcus species from pyogenic (S. agalactiae, S. pyogenes) and mitis groups (S. pneumoniae).
Collapse
Affiliation(s)
- P S Lannes-Costa
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - J S S de Oliveira
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - G da Silva Santos
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| | - P E Nagao
- Laboratory of Molecular Biology and Physiology of Streptococci, Institute of Biology Roberto Alcantara Gomes, Rio de Janeiro State University (UERJ), Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
74
|
Domon H, Terao Y. The Role of Neutrophils and Neutrophil Elastase in Pneumococcal Pneumonia. Front Cell Infect Microbiol 2021; 11:615959. [PMID: 33796475 PMCID: PMC8008068 DOI: 10.3389/fcimb.2021.615959] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Streptococcus pneumoniae, also known as pneumococcus, is a Gram-positive diplococcus and a major human pathogen. This bacterium is a leading cause of bacterial pneumonia, otitis media, meningitis, and septicemia, and is a major cause of morbidity and mortality worldwide. To date, studies on S. pneumoniae have mainly focused on the role of its virulence factors including toxins, cell surface proteins, and capsules. However, accumulating evidence indicates that in addition to these studies, knowledge of host factors and host-pathogen interactions is essential for understanding the pathogenesis of pneumococcal diseases. Recent studies have demonstrated that neutrophil accumulation, which is generally considered to play a critical role in host defense during bacterial infections, can significantly contribute to lung injury and immune subversion, leading to pneumococcal invasion of the bloodstream. Here, we review bacterial and host factors, focusing on the role of neutrophils and their elastase, which contribute to the progression of pneumococcal pneumonia.
Collapse
Affiliation(s)
- Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
75
|
Dong Y, Jin C, Ding Z, Zhu Y, He Q, Zhang X, Ai R, Yin Y, He Y. TLR4 regulates ROS and autophagy to control neutrophil extracellular traps formation against Streptococcus pneumoniae in acute otitis media. Pediatr Res 2021; 89:785-794. [PMID: 32438368 DOI: 10.1038/s41390-020-0964-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 02/29/2020] [Accepted: 04/29/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Otitis media (OM), a prevalent pediatric infectious disease, is mainly caused by Streptococcus pneumoniae (S.pn). Neutrophil extracellular traps (NETs), a novel antimicrobial strategy, were reported in 2004. We found that NETs formed in the middle ear with acute otitis media (AOM) induced by S.pn. However, the mechanisms of NETs formation are not entirely clear. METHODS We stimulated neutrophils isolated from mouse bone marrow with S.pn clinical stain 19F in vitro, and established mouse model of AOM via transbullar injection with S.pn. NETs formation, reactive oxygen species (ROS) production, autophagy activation and bacterial load were analyzed in TLR4-/- and wild-type neutrophils stimulated in vitro with S.pn and in vivo during AOM. RESULTS We found that autophagy and ROS were required for S.pn-induced NETs formation. Moreover, TLR4 partly mediated NETs formation in response to S.pn in vitro and in vivo during AOM. We also showed that attenuated NETs formation in TLR4-/- neutrophils correlated with an impaired ROS production and autophagy activation in vitro and in vivo. In addition, both the in vivo and in vitro-produced NETs were able to engulf and kill S.pn. CONCLUSIONS TLR4 regulates ROS and autophagy to control NETs formation against S.pn in the course of AOM. IMPACT S.pn can induce NETs formation in vitro and in vivo; TLR4 regulates NETs formation by ROS and autophagy; NETs contribute to the clearance of bacteria in acute otitis media. In this study, we firstly found that autophagy and ROS were required for S.pn-induced NETs formation in the model of acute otitis media (AOM). And to some extent, TLR4 mediated NETs formation during AOM. Our research might provide a potential strategy for the treatment of otitis media.
Collapse
Affiliation(s)
- Yilin Dong
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, People's Republic of China
| | - Chunfang Jin
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, People's Republic of China.,Department of Laboratory Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, People's Republic of China
| | - Zhiqiang Ding
- School of Computer Science, Chongqing Institute of Engineering, Chongqing, People's Republic of China
| | - Yiting Zhu
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, People's Republic of China
| | - Qian He
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, People's Republic of China
| | - Xinxin Zhang
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, People's Republic of China
| | - Rongshuang Ai
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, People's Republic of China
| | - Yibing Yin
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, People's Republic of China
| | - Yujuan He
- Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
76
|
The Balance of Neutrophil Extracellular Trap Formation and Nuclease Degradation: an Unknown Role of Bacterial Coinfections in COVID-19 Patients? mBio 2021; 12:mBio.03304-20. [PMID: 33593982 PMCID: PMC8545112 DOI: 10.1128/mbio.03304-20] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is leading to public health crises worldwide. An understanding of the pathogenesis and the development of treatment strategies is of high interest. Recently, neutrophil extracellular traps (NETs) have been identified as a potential driver of severe SARS-CoV-2 infections in humans. NETs are extracellular DNA fibers released by neutrophils after contact with various stimuli and accumulate antimicrobial substances or host defense peptides. When massively released, NETs are described to contribute to immunothrombosis in acute respiratory distress syndrome and in vascular occlusions. Based on the increasing evidence that NETs contribute to severe COVID-19 cases, DNase treatment of COVID-19 patients to degrade NETs is widely discussed as a potential therapeutic strategy. Here, we discuss potential detrimental effects of NETs and their nuclease degradation, since NET fragments can boost certain bacterial coinfections and thereby increase the severity of the disease.
Collapse
|
77
|
Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of Macrophages in Staphylococcus aureus Infection. Front Immunol 2021; 11:620339. [PMID: 33542723 PMCID: PMC7850989 DOI: 10.3389/fimmu.2020.620339] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 12/02/2020] [Indexed: 12/23/2022] Open
Abstract
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Collapse
Affiliation(s)
- Grace R. Pidwill
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| | - Josie F. Gibson
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Stephen A. Renshaw
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
- The Bateson Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
- Florey Institute, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
78
|
Buckland RL, Wilson AS, Brüstle A. Quantification of Neutrophil Extracellular Traps Isolated From Mouse Tissues. ACTA ACUST UNITED AC 2021; 10:e78. [PMID: 33448704 DOI: 10.1002/cpmo.78] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
One of the most intriguing functions of neutrophils is the production of neutrophil extracellular traps (NETs), which are formed when neutrophils decondense their internal DNA and extrude it along with cytotoxic proteins in a web-like structure. This process allows neutrophils to trap and kill pathogens, and is also associated with multiple hematological and autoimmune conditions. Due to their rapid degradation, there are many challenges in accurately and specifically detecting and quantifying NETs. Microscopy is the gold standard for NET detection, but is not optimal for large-scale screening. Furthermore, methods relying on detection of free DNA or on flow cytometry-based examination of NET-associated markers can be nonspecific, time-consuming, and expensive. Here, we describe an innovative, quick, specific, and inexpensive conventional flow cytometry method for detecting neutrophils on the verge of forming NETs. These methods utilize pulse-shaped analysis (PulSA) to distinguish resting neutrophils from those with decondensed DNA, a prerequisite for NET formation. An increase in DNA-diffuse neutrophils is found in cell populations after exposure to NET-inducing stimuli, consistent with the DNA decondensation expected during neutrophil NET formation. These populations are only observed in granulocytes, validating the specificity of this method. We describe protocols optimized for neutrophils retrieved from mouse blood, spleen, and bone marrow. The relative speed and simplicity of the method described here makes it a useful tool for detecting NET formation in large-scale experiments. © 2020 Wiley Periodicals LLC. Basic Protocol: Detection of nuclear decondensation in neutrophils from stimulated murine bone marrow Alternate Protocol 1: Detection of nuclear decondensation in neutrophils from splenocytes Alternate Protocol 2: Detection of nuclear decondensation in neutrophils from blood Support Protocol 1: Cryopreservation and defrosting of samples Support Protocol 2: Paraformaldehyde fixation of samples.
Collapse
Affiliation(s)
- Rebecca L Buckland
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Alicia S Wilson
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Anne Brüstle
- John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
79
|
Ulfig A, Leichert LI. The effects of neutrophil-generated hypochlorous acid and other hypohalous acids on host and pathogens. Cell Mol Life Sci 2021; 78:385-414. [PMID: 32661559 PMCID: PMC7873122 DOI: 10.1007/s00018-020-03591-y] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Neutrophils are predominant immune cells that protect the human body against infections by deploying sophisticated antimicrobial strategies including phagocytosis of bacteria and neutrophil extracellular trap (NET) formation. Here, we provide an overview of the mechanisms by which neutrophils kill exogenous pathogens before we focus on one particular weapon in their arsenal: the generation of the oxidizing hypohalous acids HOCl, HOBr and HOSCN during the so-called oxidative burst by the enzyme myeloperoxidase. We look at the effects of these hypohalous acids on biological systems in general and proteins in particular and turn our attention to bacterial strategies to survive HOCl stress. HOCl is a strong inducer of protein aggregation, which bacteria can counteract by chaperone-like holdases that bind unfolding proteins without the need for energy in the form of ATP. These chaperones are activated by HOCl through thiol oxidation (Hsp33) or N-chlorination of basic amino acid side-chains (RidA and CnoX) and contribute to bacterial survival during HOCl stress. However, neutrophil-generated hypohalous acids also affect the host system. Recent studies have shown that plasma proteins act not only as sinks for HOCl, but get actively transformed into modulators of the cellular immune response through N-chlorination. N-chlorinated serum albumin can prevent aggregation of proteins, stimulate immune cells, and act as a pro-survival factor for immune cells in the presence of cytotoxic antigens. Finally, we take a look at the emerging role of HOCl as a potential signaling molecule, particularly its role in neutrophil extracellular trap formation.
Collapse
Affiliation(s)
- Agnes Ulfig
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry-Microbial Biochemistry, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Lars I Leichert
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry-Microbial Biochemistry, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
80
|
Peng C, Li Z, Yu X. The Role of Pancreatic Infiltrating Innate Immune Cells in Acute Pancreatitis. Int J Med Sci 2021; 18:534-545. [PMID: 33390823 PMCID: PMC7757151 DOI: 10.7150/ijms.51618] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
Acute pancreatitis (AP) is a leading cause of gastrointestinal-related hospital admissions with significant morbidity and mortality. Although the underlying pathophysiology of AP is rather complex, which greatly limits the treatment options, more and more studies have revealed that infiltrating immune cells play a critical role in the pathogenesis of AP and determine disease severity. Thus, immunomodulatory therapy targeting immune cells and related inflammatory mediators is expected to be a novel treatment modality for AP which may improve the prognosis of patients. Cells of the innate immune system, including macrophages, neutrophils, dendritic cells, and mast cells, represent the majority of infiltrating cells during AP. In this review, an overview of different populations of innate immune cells and their role during AP will be discussed, with a special focus on neutrophils and macrophages.
Collapse
Affiliation(s)
- Cheng Peng
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Zhiqiang Li
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| | - Xiao Yu
- Department of Hepatopancreatobiliary Surgery, Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China
| |
Collapse
|
81
|
Yamaguchi M. [Investigation of pneumococcal virulence factors in the infection process]. Nihon Saikingaku Zasshi 2020; 75:173-183. [PMID: 33361653 DOI: 10.3412/jsb.75.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This review summarizes current knowledge regarding the pathological mechanism of Streptococcus pneumoniae, a major cause of pneumonia, sepsis, and meningitis, with focus on our previously presented studies.To identify pneumococcal adhesins or invasins on cell surfaces, we investigated several proteins with an LPXTG anchoring motif and identified one showing interaction with human fibronectin, which was designated PfbA. Next, the mechanism of pneumococcal evasion form host immunity system in blood was examined and pneumococcal α-Enolase was found to function as a neutrophil extracellular trap induction factor. Although S. pneumoniae organisms are partially killed by iron ion-induced free radicals, they have an ability to invade red blood cells and then evade antibiotics, neutrophil phagocytosis, and H2O2 killing. In addition, our findings have indicated that zinc metalloprotease ZmpC suppresses pneumococcal virulence by inhibiting bacterial invasion of the central nervous system. Since evolutionarily conserved virulence factors are potential candidate therapeutic targets, we performed molecular evolutionary analyses, which revealed that cbpJ had the highest rate of codons under negative selection to total number of codons among genes encoding choline-binding proteins. Our experimental analysis results indicated that CbpJ functions as a virulence factor in pneumococcal pneumonia by contributing to evasion of neutrophil killing.Use of a molecular biological approach based on bacterial genome sequences, clinical disease states, and molecular evolutionary analysis is an effective strategy for revealing virulence factors and important therapeutic targets.
Collapse
Affiliation(s)
- Masaya Yamaguchi
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry
| |
Collapse
|
82
|
Williams TL, Rada B, Tandon E, Gestal MC. "NETs and EETs, a Whole Web of Mess". Microorganisms 2020; 8:E1925. [PMID: 33291570 PMCID: PMC7761834 DOI: 10.3390/microorganisms8121925] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Neutrophils and eosinophils are granulocytes that have very distinct functions. Neutrophils are first responders to external threats, and they use different mechanisms to control pathogens. Phagocytosis, reactive oxygen species, and neutrophil extracellular traps (NETs) are some of the mechanisms that neutrophils utilize to fight pathogens. Although there is some controversy as to whether NETs are in fact beneficial or detrimental to the host, it mainly depends on the biological context. NETs can contribute to disease pathogenesis in certain types of diseases, while they are also undeniably critical components of the innate immune response. On the contrary, the role of eosinophils during host immune responses remains to be better elucidated. Eosinophils play an important role during helminthic infections and allergic responses. Eosinophils can function as effector cells in viral respiratory infections, gut bacterial infections, and as modulators of immune responses by driving the balance between Th1 and Th2 responses. In particular, eosinophils have biological activities that appear to be quite similar to those of neutrophils. Both possess bactericidal activity, can activate proinflammatory responses, can modulate adaptive immune responses, can form extracellular traps, and can be beneficial or detrimental to the host according to the underlying pathology. In this review we compare these two cell types with a focus on highlighting their numerous similarities related to extracellular traps.
Collapse
Affiliation(s)
- Tyler L. Williams
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| | - Balázs Rada
- Department of Infectious Diseases, University of Georgia, Athens, GA 30302, USA;
| | - Eshaan Tandon
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| | - Monica C. Gestal
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| |
Collapse
|
83
|
Ríos-López AL, González GM, Hernández-Bello R, Sánchez-González A. Avoiding the trap: Mechanisms developed by pathogens to escape neutrophil extracellular traps. Microbiol Res 2020; 243:126644. [PMID: 33199088 DOI: 10.1016/j.micres.2020.126644] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/28/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022]
Abstract
Neutrophils are the first cells of the innate immune system that respond to infection by arriving at sites when pathogens have exceeded physical barriers. Among their response mechanisms against pathogens is the release of neutrophil extracellular traps (NETs), which are composed of deoxyribonucleic acid and antimicrobial proteins such as neutrophil elastase, myeloperoxidase, antimicrobial peptides, and other proteins in neutrophil granules. The formation of extracellular traps is considered an effective strategy to capture and, in some cases, neutralize pathogenic bacteria, fungi, parasites, or viruses. However, it is also known that pathogens can respond to NETs by expressing some virulence factors, thus evading the antimicrobial effect of these structures. These include the secretion of proteins to degrade the deoxyribonucleic acid scaffold, the formation of biofilms that impede the effect of NETs, or the modification of its membrane structure to avoid interaction with NETs. In this review, we discuss these mechanisms and summarize the different pathogens that employ one or more mechanisms to evade the NET-mediated neutrophil response.
Collapse
Affiliation(s)
- A L Ríos-López
- Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, 64460, Mexico
| | - G M González
- Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, 64460, Mexico
| | - R Hernández-Bello
- Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, 64460, Mexico
| | - A Sánchez-González
- Departamento de Microbiología, Facultad de Medicina y Hospital Universitario "Dr. Jose Eleuterio Gonzalez", Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, 64460, Mexico.
| |
Collapse
|
84
|
Rosazza T, Warner J, Sollberger G. NET formation - mechanisms and how they relate to other cell death pathways. FEBS J 2020; 288:3334-3350. [PMID: 33047496 DOI: 10.1111/febs.15589] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/29/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022]
Abstract
Cell death is an integral part of both infectious and sterile inflammatory reactions. Many cell death pathways cause the dying cell to lyse, thereby amplifying inflammation. A special form of lytic cell death is the formation of neutrophil extracellular traps (NETs), large structures of chromatin and antimicrobial proteins, which are released by dying neutrophils to capture extracellular pathogens and limit the spread of infections. The molecular mechanisms of NET formation remain incompletely understood. Recent research demonstrated substantial crosstalk between different cell death pathways, most notably between apoptosis, pyroptosis and necroptosis. Here, we review suicidal and vital NET formation and discuss potential crosstalk of their mechanisms of release with other forms of cell death.
Collapse
Affiliation(s)
- Thibault Rosazza
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, UK
| | - Jordan Warner
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, UK
| | - Gabriel Sollberger
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, UK
| |
Collapse
|
85
|
Ng J, Guo F, Marneth AE, Ghanta S, Kwon MY, Keegan J, Liu X, Wright KT, Kamaz B, Cahill LA, Mullally A, Perrella MA, Lederer JA. Augmenting emergency granulopoiesis with CpG conditioned mesenchymal stromal cells in murine neutropenic sepsis. Blood Adv 2020; 4:4965-4979. [PMID: 33049055 PMCID: PMC7556132 DOI: 10.1182/bloodadvances.2020002556] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/02/2020] [Indexed: 12/17/2022] Open
Abstract
Patients with immune deficiencies from cancers and associated treatments represent a growing population within the intensive care unit with increased risk of morbidity and mortality from sepsis. Mesenchymal stromal cells (MSCs) are an integral part of the hematopoietic niche and express toll-like receptors, making them candidate cells to sense and translate pathogenic signals into an innate immune response. In this study, we demonstrate that MSCs administered therapeutically in a murine model of radiation-associated neutropenia have dual actions to confer a survival benefit in Pseudomonas aeruginosa pneumo-sepsis that is not from improved bacterial clearance. First, MSCs augment the neutrophil response to infection, an effect that is enhanced when MSCs are preconditioned with CpG oligodeoxynucleotide, a toll-like receptor 9 agonist. Using cytometry by time of flight, we identified proliferating neutrophils (Ly6GlowKi-67+) as the main expanded cell population within the bone marrow. Further analysis revealed that CpG-MSCs expand a lineage restricted progenitor population (Lin-Sca1+C-kit+CD150-CD48+) in the bone marrow, which corresponded to a doubling in the myeloid proliferation and differentiation potential in response to infection compared with control. Despite increased neutrophils, no reduction in organ bacterial count was observed between experimental groups. However, the second effect exerted by CpG-MSCs is to attenuate organ damage, particularly in the lungs. Neutrophils obtained from irradiated mice and cocultured with CpG-MSCs had decreased neutrophil extracellular trap formation, which was associated with decreased citrullinated H3 staining in the lungs of mice given CpG-MSCs in vivo. Thus, this preclinical study provides evidence for the therapeutic potential of MSCs in neutropenic sepsis.
Collapse
Affiliation(s)
- Julie Ng
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | | | | | - Min-Young Kwon
- Division of Pulmonary and Critical Care, Department of Medicine
| | | | - Xiaoli Liu
- Division of Pulmonary and Critical Care, Department of Medicine
- Department of Pediatric Newborn Medicine, and
| | - Kyle T Wright
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | | | | | - Mark A Perrella
- Division of Pulmonary and Critical Care, Department of Medicine
- Department of Pediatric Newborn Medicine, and
| | | |
Collapse
|
86
|
Feldman C, Anderson R. Platelets and Their Role in the Pathogenesis of Cardiovascular Events in Patients With Community-Acquired Pneumonia. Front Immunol 2020; 11:577303. [PMID: 33042161 PMCID: PMC7527494 DOI: 10.3389/fimmu.2020.577303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Community-acquired pneumonia (CAP) remains an important cause of morbidity and mortality throughout the world with much recent and ongoing research focused on the occurrence of cardiovascular events (CVEs) during the infection, which are associated with adverse short-term and long-term survival. Much of the research directed at unraveling the pathogenesis of these events has been undertaken in the settings of experimental and clinical CAP caused by the dangerous, bacterial respiratory pathogen, Streptococcus pneumoniae (pneumococcus), which remains the most common bacterial cause of CAP. Studies of this type have revealed that although platelets play an important role in host defense against infection, there is also increasing recognition that hyperactivation of these cells contributes to a pro-inflammatory, prothrombotic systemic milieu that contributes to the etiology of CVEs. In the case of the pneumococcus, platelet-driven myocardial damage and dysfunction is exacerbated by the direct cardiotoxic actions of pneumolysin, a major pore-forming toxin of this pathogen, which also acts as potent activator of platelets. This review is focused on the role of platelets in host defense against infection, including pneumococcal infection in particular, and reviews the current literature describing the potential mechanisms by which platelet activation contributes to cardiovascular complications in CAP. This is preceded by an evaluation of the burden of pneumococcal infection in CAP, the clinical features and putative pathogenic mechanisms of the CVE, and concludes with an evaluation of the potential utility of the anti-platelet activity of macrolides and various adjunctive therapies.
Collapse
Affiliation(s)
- Charles Feldman
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, Institute of Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
87
|
Santocki M, Kolaczkowska E. On Neutrophil Extracellular Trap (NET) Removal: What We Know Thus Far and Why So Little. Cells 2020; 9:cells9092079. [PMID: 32932841 PMCID: PMC7565917 DOI: 10.3390/cells9092079] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/05/2020] [Accepted: 09/08/2020] [Indexed: 12/17/2022] Open
Abstract
Although neutrophil extracellular traps (NETs) were discovered only 16 years ago, they have already taken us from heaven to hell as we learned that apart from beneficial trapping of pathogens, they cause, or contribute to, numerous disorders. The latter is connected to their persistent presence in the blood or tissue, and we hardly know how they are removed in mild pathophysiological conditions and why their removal is impaired in multiple severe pathological conditions. Herein, we bring together all data available up till now on how NETs are cleared—from engaged cells, their phenotypes, to involved enzymes and molecules. Moreover, we hypothesize on why NET removal is challenged in multiple disorders and propose further directions for studies on NET removal as well as possible therapeutic strategies to have them cleared.
Collapse
|
88
|
Soh KY, Loh JMS, Hall C, Proft T. Functional Analysis of Two Novel Streptococcus iniae Virulence Factors Using a Zebrafish Infection Model. Microorganisms 2020; 8:E1361. [PMID: 32899555 PMCID: PMC7564053 DOI: 10.3390/microorganisms8091361] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 08/31/2020] [Accepted: 09/03/2020] [Indexed: 12/30/2022] Open
Abstract
Streptococcus iniae is a major fish pathogen that contributes to large annual losses in the aquaculture industry, exceeding US$100 million. It is also reported to cause opportunistic infections in humans. We have recently identified two novel S. iniae virulence factors, an extracellular nuclease (SpnAi) and a secreted nucleotidase (S5nAi), and verified their predicted enzymatic activities using recombinant proteins. Here, we report the generation of green fluorescent S. iniae spnAi and s5nAi deletion mutants and their evaluation in a transgenic zebrafish infection model. Our results show nuclease and nucleotidase activities in S. iniae could be attributed to SpnAi and S5nAi, respectively. Consistent with this, larvae infected with the deletion mutants demonstrated enhanced survival and bacterial clearance, compared to those infected with wild-type (WT) S. iniae. Deletion of spnAi and s5nAi resulted in sustained recruitment of neutrophils and macrophages, respectively, to the site of infection. We also show that recombinant SpnAi is able to degrade neutrophil extracellular traps (NETs) isolated from zebrafish kidney tissue. Our results suggest that both enzymes play an important role in S. iniae immune evasion and might present potential targets for the development of therapeutic agents or vaccines.
Collapse
Affiliation(s)
- Kar Yan Soh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland 1142, New Zealand; (K.Y.S.); (J.M.S.L.)
- Maurice Wilkins Centre for Biomolecular Discoveries, The University of Auckland, Auckland 1142, New Zealand
| | - Jacelyn Mei San Loh
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland 1142, New Zealand; (K.Y.S.); (J.M.S.L.)
- Maurice Wilkins Centre for Biomolecular Discoveries, The University of Auckland, Auckland 1142, New Zealand
| | - Christopher Hall
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland 1142, New Zealand; (K.Y.S.); (J.M.S.L.)
| | - Thomas Proft
- Department of Molecular Medicine & Pathology, School of Medical Sciences, The University of Auckland, Auckland 1142, New Zealand; (K.Y.S.); (J.M.S.L.)
- Maurice Wilkins Centre for Biomolecular Discoveries, The University of Auckland, Auckland 1142, New Zealand
| |
Collapse
|
89
|
Burgener SS, Schroder K. Neutrophil Extracellular Traps in Host Defense. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037028. [PMID: 31767647 DOI: 10.1101/cshperspect.a037028] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neutrophils are produced in the bone marrow and then patrol blood vessels from which they can be rapidly recruited to a site of infection. Neutrophils bind, engulf, and efficiently kill invading microbes via a suite of defense mechanisms. Diverse extracellular and intracellular microbes induce neutrophils to extrude neutrophil extracellular traps (NETs) through the process of NETosis. Here, we review the signaling mechanisms and cell biology underpinning the key NETosis pathways during infection and the antimicrobial functions of NETs in host defense.
Collapse
Affiliation(s)
- Sabrina Sofia Burgener
- Institute for Molecular Bioscience (IMB), and IMB Centre for Inflammation and Disease Research, The University of Queensland, St Lucia 4072, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), and IMB Centre for Inflammation and Disease Research, The University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
90
|
Demarco B, Chen KW, Broz P. Cross talk between intracellular pathogens and cell death. Immunol Rev 2020; 297:174-193. [PMID: 32567717 DOI: 10.1111/imr.12892] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/24/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023]
Abstract
Infections with bacterial pathogens often results in the initiation of programmed cell death as part of the host innate immune defense, or as a bacterial virulence strategy. Induction of host cell death is controlled by an elaborate network of innate immune and cell death signaling pathways and manifests in different morphologically and functionally distinct forms of death, such as apoptosis, necroptosis, NETosis and pyroptosis. The mechanism by which host cell death restricts bacterial replication is highly cell-type and context depended, but its physiological importance is highlighted the diversity of strategies bacterial pathogens use to avoid induction of cell death or to block cell death signaling pathways. In this review, we discuss the latest insights into how bacterial pathogens elicit and manipulate cell death signaling, how different forms of cell death kill or restrict bacteria and how cell death and innate immune pathway cross talk to guard against pathogen-induced inhibition of host cell death.
Collapse
Affiliation(s)
- Benjamin Demarco
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Kaiwen W Chen
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Petr Broz
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
91
|
Magán-Fernández A, Rasheed Al-Bakri SM, O’Valle F, Benavides-Reyes C, Abadía-Molina F, Mesa F. Neutrophil Extracellular Traps in Periodontitis. Cells 2020; 9:cells9061494. [PMID: 32575367 PMCID: PMC7349145 DOI: 10.3390/cells9061494] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 02/08/2023] Open
Abstract
Neutrophils are key cells of the immune system and have a decisive role in fighting foreign pathogens in infectious diseases. Neutrophil extracellular traps (NETs) consist of a mesh of DNA enclosing antimicrobial peptides and histones that are released into extracellular space following neutrophil response to a wide range of stimuli, such as pathogens, host-derived mediators and drugs. Neutrophils can remain functional after NET formation and are important for periodontal homeostasis. Periodontitis is an inflammatory multifactorial disease caused by a dysbiosis state between the gingival microbiome and the immune response of the host. The pathogenesis of periodontitis includes an immune-inflammatory component in which impaired NET formation and/or elimination can be involved, contributing to an exacerbated inflammatory reaction and to the destruction of gingival tissue. In this review, we summarize the current knowledge about the role of NETs in the pathogenesis of periodontitis.
Collapse
Affiliation(s)
- Antonio Magán-Fernández
- Periodontology Department, School of Dentistry, University of Granada, 18071 Granada, Spain; (A.M.-F.); (S.M.R.A.-B.); (F.M.)
| | - Sarmad Muayad Rasheed Al-Bakri
- Periodontology Department, School of Dentistry, University of Granada, 18071 Granada, Spain; (A.M.-F.); (S.M.R.A.-B.); (F.M.)
| | - Francisco O’Valle
- Pathology Department, School of Medicine (IBIMER, CIBM), University of Granada, 18071 Granada, Spain;
- Biosanitary Research Institute (IBS-GRANADA), University of Granada, 18012 Granada, Spain
| | - Cristina Benavides-Reyes
- Department of Operative Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Correspondence: ; Tel.: +34-9-5824-0654
| | - Francisco Abadía-Molina
- Department of Cell Biology, University of Granada, 18071 Granada, Spain;
- INYTA, Institute of Nutrition and Food Technology “José Mataix”, University of Granada, Armilla, 18100 Granada, Spain
| | - Francisco Mesa
- Periodontology Department, School of Dentistry, University of Granada, 18071 Granada, Spain; (A.M.-F.); (S.M.R.A.-B.); (F.M.)
| |
Collapse
|
92
|
|
93
|
Yousefi S, Simon D, Stojkov D, Karsonova A, Karaulov A, Simon HU. In vivo evidence for extracellular DNA trap formation. Cell Death Dis 2020; 11:300. [PMID: 32355207 PMCID: PMC7193637 DOI: 10.1038/s41419-020-2497-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/01/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022]
Abstract
Extracellular DNA trap formation is a cellular function of neutrophils, eosinophils, and basophils that facilitates the immobilization and killing of invading microorganisms in the extracellular milieu. To form extracellular traps, granulocytes release a scaffold consisting of mitochondrial DNA in association with granule proteins. As we understand more about the molecular mechanism for the formation of extracellular DNA traps, the in vivo function of this phenomenon under pathological conditions remains an enigma. In this article, we critically review the literature to summarize the evidence for extracellular DNA trap formation under in vivo conditions. Extracellular DNA traps have not only been detected in infectious diseases but also in chronic inflammatory diseases, as well as in cancer. While on the one hand, extracellular DNA traps clearly exhibit an important function in host defense, it appears that they can also contribute to the maintenance of inflammation and metastasis, suggesting that they may represent an interesting drug target for such pathological conditions.
Collapse
Affiliation(s)
- Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Antonina Karsonova
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia.
| |
Collapse
|
94
|
Disruption of the cpsE and endA Genes Attenuates Streptococcus pneumoniae Virulence: Towards the Development of a Live Attenuated Vaccine Candidate. Vaccines (Basel) 2020; 8:vaccines8020187. [PMID: 32326482 PMCID: PMC7349068 DOI: 10.3390/vaccines8020187] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 03/27/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
The majority of deaths due to Streptococcus pneumoniae infections are in developing countries. Although polysaccharide-based pneumococcal vaccines are available, newer types of vaccines are needed to increase vaccine affordability, particularly in developing countries, and to provide broader protection across all pneumococcal serotypes. To attenuate pneumococcal virulence with the aim of engineering candidate live attenuated vaccines (LAVs), we constructed knockouts in S. pneumoniae D39 of one of the capsular biosynthetic genes, cpsE that encodes glycosyltransferase, and the endonuclease gene, endA, that had been implicated in the uptake of DNA from the environment as well as bacterial escape from neutrophil-mediated killing. The cpsE gene knockout significantly lowered peak bacterial density, BALB/c mice nasopharyngeal (NP) colonisation but increased biofilm formation when compared to the wild-type D39 strain as well as the endA gene knockout mutant. All constructed mutant strains were able to induce significantly high serum and mucosal antibody response in BALB/c mice. However, the cpsE-endA double mutant strain, designated SPEC, was able to protect mice from high dose mucosal challenge of the D39 wild-type. Furthermore, SPEC showed 23-fold attenuation of virulence compared to the wild-type. Thus, the cpsE-endA double-mutant strain could be a promising candidate for further development of a LAV for S. pneumoniae.
Collapse
|
95
|
Abrams ST, Morton B, Alhamdi Y, Alsabani M, Lane S, Welters ID, Wang G, Toh CH. A Novel Assay for Neutrophil Extracellular Trap Formation Independently Predicts Disseminated Intravascular Coagulation and Mortality in Critically Ill Patients. Am J Respir Crit Care Med 2020; 200:869-880. [PMID: 31162936 DOI: 10.1164/rccm.201811-2111oc] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Rationale: Neutrophil extracellular traps (NETs) are important in the host defense against infection, but they also promote intravascular coagulation and multiorgan failure in animal models. Their clinical significance remains unclear, and available assays for patient care lack specificity and reliability.Objectives: To establish a novel assay and test its clinical significance.Methods: A prospective cohort of 341 consecutive adult ICU patients was recruited. The NET-forming capacity of ICU admission blood samples was semiquantified by directly incubating patient plasma with isolated neutrophils ex vivo. The association of NET-forming capacity with Sequential Organ Failure Assessment scores, disseminated intravascular coagulation, and 28-day mortality was analyzed and compared with available NET assays.Measurements and Main Results: Using the novel assay, we could stratify ICU patients into four groups with absent (22.0%), mild (49.9%), moderate (14.4%), and strong (13.8%) NET formation, respectively. Strong NET formation was predominantly found in sepsis (P < 0.0001). Adjusted by Acute Physiology and Chronic Health Evaluation II score, multivariate regression showed that the degree of NET formation could independently predict disseminated intravascular coagulation and mortality, whereas other NET assays (e.g., cell-free DNA, myeloperoxidase, and myeloperoxidase-DNA complexes) could not. IL-8 concentrations were found to be strongly associated with NET formation, and inhibiting IL-8 significantly attenuated NETosis. Mitogen-activated protein kinase activation by IL-8 has been identified as a major pathway of NET formation in patients.Conclusions: This assay directly measures the NET-forming capacity in patient plasma. This could guide clinical management and enable identification of NET-inducing factors in individual patients for targeted treatment and personalized ICU medicine.
Collapse
Affiliation(s)
| | - Ben Morton
- Aintree University Hospital NHS Foundation Trust, Liverpool, United Kingdom.,Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom; and
| | | | | | | | - Ingeborg D Welters
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, United Kingdom.,Intensive Care Unit and
| | | | - Cheng-Hock Toh
- Institute of Infection and Global Health.,Roald Dahl Haemostasis & Thrombosis Centre, Royal Liverpool University Hospital, Liverpool, United Kingdom
| |
Collapse
|
96
|
Shi Y, Liu T, Nieman DC, Cui Y, Li F, Yang L, Shi H, Chen P. Aerobic Exercise Attenuates Acute Lung Injury Through NET Inhibition. Front Immunol 2020; 11:409. [PMID: 32265910 PMCID: PMC7096358 DOI: 10.3389/fimmu.2020.00409] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction: Aerobic exercise improves lung inflammation in acute lung injury (ALI), but its mechanism remains unknown. Neutrophil extracellular traps (NETs) play an important role in LPS-induced ALI, and a positive correlation exists between NET formation and proinflammatory macrophage polarization. This study investigated whether aerobic exercise reduces the pro-inflammatory polarization of alveolar macrophages (AMs) by inhibiting the excessive release of NETs and then alleviating the inflammatory response of ALI. Methods: C57BL/6 male mice were randomly divided into four groups: sedentary group (CON), sedentary and extra-pulmonary LPS injection group (LPS), 5-weeks aerobic training intervention and LPS injection group (EXE+LPS), and DNase I plus LPS injection group (DNase+LPS). Twenty-four hours after drug injection, bronchoalveolar lavage fluid (BALF), AM, and lung tissues were obtained to detect inflammatory responses, NET formation, macrophage polarization, and protein activation. In the in vitro study, a murine AM cell line, designated MH-S, was stimulated with LPS, purified NETs, and NETs plus DNase I. Results: EXE+LPS and DNase+LPS mice exhibited reduced neutrophil infiltration, decreased NET release, and lower pro-inflammatory polarization of AM compared with LPS mice. Subsequently, Western blot showed inhibition of the phosphorylation of MAPK and NF-κB proteins of AMs in EXE+LPS and DNase+LPS mice compared with LPS mice. Lastly, stimulation of MH-S cells by NETs revealed a trend for pro-inflammatory cell polarization, with NF-κB protein activation at 8 h and ERK1/2 activation at 1, 2, and 8 h. Conclusions: Aerobic exercise alleviated ALI through NET-induced AM pro-inflammatory polarization involving ERK1/2 and NF-κB signaling.
Collapse
Affiliation(s)
- Yue Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Tingting Liu
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - David C Nieman
- North Carolina Research Campus, Appalachian State University, Kannapolis, NC, United States
| | - Yanqiu Cui
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Fei Li
- School of Physical Education and Sport Training, Shanghai University of Sport, Shanghai, China
| | - Luyu Yang
- Department of General Surgery, Cancer Metastasis Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Hui Shi
- Department of Rheumatology and Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peijie Chen
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
97
|
Bacterial non-specific nucleases of the phospholipase D superfamily and their biotechnological potential. Appl Microbiol Biotechnol 2020; 104:3293-3304. [PMID: 32086594 DOI: 10.1007/s00253-020-10459-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 12/19/2022]
Abstract
Bacterial non-specific nucleases are ubiquitously distributed and involved in numerous intra- and extracellular processes. Although all nucleases share the basic chemistry for the hydrolysis of phosphodiester bonds in nucleic acid molecules, the catalysis comprises diverse modes of action, which offers great potential for versatile biotechnological applications. A major criterium for their differentiation is substrate specificity. Specific endonucleases are widely used as restriction enzymes in molecular biology approaches, whereas the main applications of non-specific nucleases (NSNs) are the removal of nucleic acids from crude extracts in industrial downstream processing and the prevention of cell clumping in microfabricated channels. In nature, the predominant role of NSNs is the acquisition of nutrient sources such as nucleotides and phosphates. The number of extensively characterized NSNs and available structures is limited. Moreover, their applicability is mostly challenged by the presence of metal chelators that impede the hydrolysis of nucleic acids in a metal ion-dependent manner. However, a few metal ion-independent NSNs that tolerate the presence of metal chelators have been characterized in recent years with none being commercially available to date. The classification and biotechnological potential of bacterial NSNs with a special focus on metal ion-independent nucleases are presented and discussed.Key Points • Bacterial phospholipases (PLD-family) exhibit nucleolytic activity. • Bacterial nucleases of the PLD-family are metal ion-independent. • NSNs can be used in downstream processing approaches.
Collapse
|
98
|
Neumann A, Brogden G, von Köckritz-Blickwede M. Extracellular Traps: An Ancient Weapon of Multiple Kingdoms. BIOLOGY 2020; 9:biology9020034. [PMID: 32085405 PMCID: PMC7168307 DOI: 10.3390/biology9020034] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/31/2022]
Abstract
The discovery, in 2004, of extracellular traps released by neutrophils has extended our understanding of the mode of action of various innate immune cells. This fascinating discovery demonstrated the extracellular trapping and killing of various pathogens by neutrophils. During the last decade, evidence has accumulated showing that extracellular traps play a crucial role in the defence mechanisms of various cell types present in vertebrates, invertebrates, and plants. The aim of this review is to summarise the relevant literature on the evolutionary history of extracellular traps used as a weapon in various kingdoms of life.
Collapse
Affiliation(s)
- Ariane Neumann
- Department of Clinical Sciences, Division of Infection Medicine, Lund University, Baravägen 27, 22184 Lund, Sweden;
| | - Graham Brogden
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hannover, Germany;
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hannover, Germany;
- Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
- Correspondence: ; Tel.: +49-511-953-8787
| |
Collapse
|
99
|
Peng L, Li L, He XL, Yu JY, Zeng ZJ, Yang WJ, Zhang B, Zhang TS, Cao H, Huang SH, Liu LQ. Memantine Displays Antimicrobial Activity by Enhancing Escherichia coli Pathogen-Induced Formation of Neutrophil Extracellular Traps. Front Cell Infect Microbiol 2020; 10:47. [PMID: 32117815 PMCID: PMC7031421 DOI: 10.3389/fcimb.2020.00047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 01/24/2020] [Indexed: 01/31/2023] Open
Abstract
Bacterial infection remains one of the leading causes of death worldwide due to the continuous rise of multiple antibiotic-resistant bacteria. Focusing solely on bacteria as the drug targets is a major limitation inherent in the conventional antibiotic therapy. Recently, host-directed therapies have become such an innovative approach to modulate the host defense system and the interplay of innate and adaptive immunity. Our previous studies showed that memantine (MEM), an α7 nAChR antagonist, could efficiently block multi-drug resistant Escherichia coli-caused bacteremia and meningitis in a mouse model. However, the underlying mechanisms that govern the antibacterial effects of MEM are still unknown. In this study, we demonstrated that MEM is able to significantly suppress E. coli infection by enhancing E. coli-induced formation and release of NETs in vitro and in vivo. MEM could promote the trapping and bactericidal activities of the polymorphonuclear neutrophils (PMNs) in a manner dependent on α7 nAChR, since knockdown of this receptor noticeably reduces the survival ability of bacteria in PMNs while MEM no longer affects the survival of bacteria in PMNs. Our results also showed that when the expression of S100A9, an antiseptic protein, is inhibited, pathogen survival rates in PMNs increase significantly. MEM reverses this effect in a concentration-dependent manner. MEM stimulates the production of MPO, S100A9, and DNA in PMNs and accelerates the release of depolymerized chromatin fibers into the extracellular space, suggesting the formation of NETs. Taken together, our data suggest that MEM effectively blocks bacterial infection through the promotion of the antibacterial function of NETs induced by E. coli.
Collapse
Affiliation(s)
- Liang Peng
- Department of Clinical Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Li
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, China
- Department of Pediatrics, Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - Xiao-Long He
- Guangdong Provincial Key Laboratory of Tropical Diseases, Department of Microbiology, Southern Medical University, Guangzhou, China
| | - Jing-Yi Yu
- Department of Pediatrics, Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
| | - Zhi-Jie Zeng
- Guangdong Provincial Key Laboratory of Tropical Diseases, Department of Microbiology, Southern Medical University, Guangzhou, China
| | - Wei-Jun Yang
- Guangdong Provincial Key Laboratory of Tropical Diseases, Department of Microbiology, Southern Medical University, Guangzhou, China
| | - Bao Zhang
- Department of Pediatrics, Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
- Guangdong Provincial Key Laboratory of Tropical Diseases, Department of Microbiology, Southern Medical University, Guangzhou, China
| | - Tie-Song Zhang
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, China
| | - Hong Cao
- Guangdong Provincial Key Laboratory of Tropical Diseases, Department of Microbiology, Southern Medical University, Guangzhou, China
| | - Sheng-He Huang
- Kunming Key Laboratory of Children Infection and Immunity, Yunnan Institute of Pediatrics, Kunming Children's Hospital, Kunming, China
- Department of Pediatrics, Saban Research Institute, Childrens Hospital Los Angeles, University of Southern California, Los Angeles, CA, United States
- Guangdong Provincial Key Laboratory of Tropical Diseases, Department of Microbiology, Southern Medical University, Guangzhou, China
| | - Li-Qun Liu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
100
|
Deng Q, Pan B, Alam HB, Liang Y, Wu Z, Liu B, Mor-Vaknin N, Duan X, Williams AM, Tian Y, Zhang J, Li Y. Citrullinated Histone H3 as a Therapeutic Target for Endotoxic Shock in Mice. Front Immunol 2020; 10:2957. [PMID: 31998291 PMCID: PMC6962130 DOI: 10.3389/fimmu.2019.02957] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022] Open
Abstract
Sepsis results in millions of deaths every year, with acute lung injury (ALI) being one of the leading causes of mortality in septic patients. As neutrophil extracellular traps (NETs) are abundant in sepsis, neutralizing components of NETs may be a useful strategy to improve outcomes of sepsis. Citrullinated histone H3 (CitH3) has been recently shown to be involved in the NET formation. In this study, we demonstrate that CitH3 damages human umbilical vein endothelial cells (HUVECs) and potentiates NET formation through a positive feedback mechanism. We developed a novel CitH3 monoclonal antibody to target peptidylarginine deiminase (PAD) 2 and PAD 4 generated CitH3. In a mouse model of lethal lipopolysaccharide (LPS) induced shock, neutralizing CitH3 with the newly developed anti-CitH3 monoclonal antibody attenuates inflammatory responses, ameliorates ALI, and improves survival. Our study suggests that effectively blocking circulating CitH3 might be a potential therapeutic method for the treatment of endotoxemia.
Collapse
Affiliation(s)
- Qiufang Deng
- Xiangya Hospital, Central South University, Changsha, China.,Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Baihong Pan
- Xiangya Hospital, Central South University, Changsha, China.,Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Hasan B Alam
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Yingjian Liang
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States.,The First Hospital, China Medical University, Shenyang, China
| | - Zhenyu Wu
- Xiangya Hospital, Central South University, Changsha, China.,Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Baoling Liu
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Nirit Mor-Vaknin
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Xiuzhen Duan
- Department of Pathology, Loyola University Medical Center, Maywood, IL, United States
| | - Aaron M Williams
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Yuzi Tian
- Xiangya Hospital, Central South University, Changsha, China.,Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Justin Zhang
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| | - Yongqing Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|