51
|
Zhang Y, Wang Q, Mackay CR, Ng LG, Kwok I. Neutrophil subsets and their differential roles in viral respiratory diseases. J Leukoc Biol 2022; 111:1159-1173. [PMID: 35040189 PMCID: PMC9015493 DOI: 10.1002/jlb.1mr1221-345r] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 12/19/2022] Open
Abstract
Neutrophils play significant roles in immune homeostasis and as neutralizers of microbial infections. Recent evidence further suggests heterogeneity of neutrophil developmental and activation states that exert specialized effector functions during inflammatory disease conditions. Neutrophils can play multiple roles during viral infections, secreting inflammatory mediators and cytokines that contribute significantly to host defense and pathogenicity. However, their roles in viral immunity are not well understood. In this review, we present an overview of neutrophil heterogeneity and its impact on the course and severity of viral respiratory infectious diseases. We focus on the evidence demonstrating the crucial roles neutrophils play in the immune response toward respiratory infections, using influenza as a model. We further extend the understanding of neutrophil function with the studies pertaining to COVID-19 disease and its neutrophil-associated pathologies. Finally, we discuss the relevance of these results for future therapeutic options through targeting and regulating neutrophil-specific responses.
Collapse
Affiliation(s)
- Yuning Zhang
- Department of ResearchNational Skin CentreSingaporeSingapore
| | - Quanbo Wang
- School of Pharmaceutical Sciences, Shandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Charles R Mackay
- School of Pharmaceutical Sciences, Shandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
- Department of Microbiology, Infection and Immunity ProgramBiomedicine Discovery Institute, Monash UniversityMelbourneAustralia
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN)A*STAR (Agency for Science, Technology and Research)BiopolisSingapore
- State Key Laboratory of Experimental HematologyInstitute of Hematology, Chinese Academy of Medical Sciences & Peking Union Medical CollegeTianjinChina
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
- Department of Microbiology and ImmunologyImmunology Translational Research Program, Yong Loo Lin School of Medicine, Immunology Program, Life Sciences Institute, National University of SingaporeSingaporeSingapore
- National Cancer Centre SingaporeSingaporeSingapore
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN)A*STAR (Agency for Science, Technology and Research)BiopolisSingapore
| |
Collapse
|
52
|
Thimmappa PY, Nair AS, Najar MA, Mohanty V, Shastry S, Prasad TSK, Joshi MB. Quantitative phosphoproteomics reveals diverse stimuli activate distinct signaling pathways during neutrophil activation. Cell Tissue Res 2022; 389:241-257. [PMID: 35622142 PMCID: PMC9287233 DOI: 10.1007/s00441-022-03636-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/11/2022] [Indexed: 12/19/2022]
Abstract
Neutrophils display functional heterogeneity upon responding diversely to physiological and pathological stimulations. During type 2 diabetes (T2D), hyperglycemia constitutively activates neutrophils, leading to reduced response to infections and on the other hand, elevated metabolic intermediates such as homocysteine induce bidirectional activation of platelets and neutrophils leading to thrombosis. Hence, in the context of T2D-associated complications, we examined the influence of high glucose, homocysteine, and LPS representing effector molecules of hyperglycemia, thrombosis, and infection, respectively, on human neutrophil activation to identify distinct signaling pathways by quantitative phosphoproteomics approach. High glucose activated C-Jun-N-Terminal Kinase, NTRK1, SYK, and PRKACA kinases associated with Rho GTPase signaling and phagocytosis, whereas LPS induced AKT1, SRPK2, CSNK2A1, and TTN kinases involved in cytokine signaling and inflammatory response. Homocysteine treatment led to activatation of LRRK2, FGR, MAPK3, and PRKCD kinases which are associated with neutrophil degranulation and cytoskeletal remodeling. Diverse inducers differentially modulated phosphorylation of proteins associated with neutrophil functions such as oxidative burst, degranulation, extracellular traps, and phagocytosis. Further validation of phosphoproteomics data on selected kinases revealed neutrophils pre-cultured under high glucose showed impeded response to LPS to phosphorylate p-ERK1/2Thr202/Tyr204, p-AKTSer473, and C-Jun-N-Terminal KinaseSer63 kinases. Our study provides novel phosphoproteome signatures that may be explored to understand neutrophil biology in T2D-associated complications.
Collapse
Affiliation(s)
- Pooja Yedehalli Thimmappa
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Aswathy S Nair
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Mohd Altaf Najar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575020, India
| | - Varshasnatha Mohanty
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575020, India
| | - Shamee Shastry
- Department of Immunohematology and Blood Transfusion, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, 576104, India
| | | | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
53
|
Myeloid-Specific Pyruvate-Kinase-Type-M2-Deficient Mice Are Resistant to Acute Lung Injury. Biomedicines 2022; 10:biomedicines10051193. [PMID: 35625931 PMCID: PMC9138865 DOI: 10.3390/biomedicines10051193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 02/05/2023] Open
Abstract
Infiltration of polymorphonuclear neutrophils (PMNs) plays a central role in acute lung injury (ALI). The mechanisms governing PMN inflammatory responses, however, remain incompletely understood. Based on our recent study showing a non-metabolic role of pyruvate kinase type M2 (PKM2) in controlling PMN degranulation of secondary and tertiary granules and consequent chemotaxis, here we tested a hypothesis that Pkm2-deficient mice may resist ALI due to impaired PMN inflammatory responses. We found that PMN aerobic glycolysis controlled the degranulation of secondary and tertiary granules induced by fMLP and PMA. Compared to WT PMNs, Pkm2-deficient (Pkm2-/-) PMNs displayed significantly less capacity for fMLP- or PMA-induced degranulation of secondary and tertiary granules, ROS production, and transfilter migration. In line with this, myeloid-specific Pkm2-/- mice exhibited impaired zymosan-induced PMN infiltration in the peritoneal cavity. Employing an LPS-induced ALI mouse model, LPS-treated Pkm2-/- mice displayed significantly less infiltration of inflammatory PMNs in the alveolar space and a strong resistance to LPS-induced ALI. Our results thus reveal that PKM2 is required for PMN inflammatory responses and deletion of PKM2 in PMN leads to an impaired PMN function but protection against LPS-induced ALI.
Collapse
|
54
|
Polak D, Bohle B. Neutrophils-typical atypical antigen presenting cells? Immunol Lett 2022; 247:52-58. [DOI: 10.1016/j.imlet.2022.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 11/05/2022]
|
55
|
Wang G, Nauseef WM. Neutrophil dysfunction in the pathogenesis of cystic fibrosis. Blood 2022; 139:2622-2631. [PMID: 35213685 PMCID: PMC9053701 DOI: 10.1182/blood.2021014699] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/31/2022] [Indexed: 11/20/2022] Open
Abstract
Polymorphonuclear neutrophils (PMNs) figure prominently in host defense against infection and in noninfectious inflammation. Mobilized early in an inflammatory response, PMNs mediate immediate cellular defense against microbes and orchestrate events that culminate in cessation of inflammation and restoration of homeostasis. Failure to terminate the inflammatory response and its causes can fuel exuberant inflammation characteristic of many human diseases, including cystic fibrosis (CF), an autosomal recessive genetic disease caused by mutations in the CF transmembrane conductance regulator. CF affects multiple end organs, with persistent bacterial infection and chronic neutrophilic inflammation in airways predominating the clinical picture. To match the diverse microbial challenges that they may encounter, PMNs possess a variety of antimicrobial systems to slow or kill invading microorganisms confined in their phagosomes. Prominent among PMN defense systems is their ability to generate hypochlorous acid, a potent microbicide, by reacting oxidants generated by the NADPH oxidase with myeloperoxidase (MPO) released from azurophilic granules in the presence of chloride (Cl-). Products of the MPO-H2O2-Cl system oxidize susceptible biomolecules and support robust antimicrobial action against many, but not all, potential human pathogens. Underscoring that the MPO-H2O2-Cl system is integral to optimal host defense and proper regulation of inflammation, individuals with defects in any component of this system, as seen in chronic granulomatous disease or MPO deficiency, incur increased rates or severity of infection and signs of dysregulated inflammatory responses. We focus attention in this review on the molecular basis for and the clinical consequences of defects in the MPO-H2O2-Cl system because of the compromised Cl transport seen in CF. We will discuss first how the MPO-H2O2-Cl system in healthy PMNs participates in host defense and resolution of inflammation and then review how a defective MPO-H2O2-Cl system contributes to the increased susceptibility to infection and dysregulated inflammation associated with the clinical manifestations of CF.
Collapse
Affiliation(s)
- Guoshun Wang
- Department of Microbiology, Immunology, and Parasitology, and
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA
| | - William M Nauseef
- Inflammation Program, Department of Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA; and
- Veterans Administration Medical Center, Iowa City, IA
| |
Collapse
|
56
|
Zhang N, Aiyasiding X, Li WJ, Liao HH, Tang QZ. Neutrophil degranulation and myocardial infarction. Cell Commun Signal 2022; 20:50. [PMID: 35410418 PMCID: PMC8996539 DOI: 10.1186/s12964-022-00824-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 12/26/2021] [Indexed: 11/21/2022] Open
Abstract
Myocardial infarction (MI) is one of the most common cardiac emergencies with high morbidity and is a leading cause of death worldwide. Since MI could develop into a life-threatening emergency and could also seriously affect the life quality of patients, continuous efforts have been made to create an effective strategy to prevent the occurrence of MI and reduce MI-related mortality. Numerous studies have confirmed that neutrophils play important roles in inflammation and innate immunity, which provide the first line of defense against microorganisms by producing inflammatory cytokines and chemokines, releasing reactive oxygen species, and degranulating components of neutrophil cytoplasmic granules to kill pathogens. Recently, researchers reported that neutrophils are closely related to the severity and prognosis of patients with MI, and neutrophil to lymphocyte ratio in post-MI patients had predictive value for major adverse cardiac events. Neutrophils have been increasingly recognized to exert important functions in MI. Especially, granule proteins released by neutrophil degranulation after neutrophil activation have been suggested to involve in the process of MI. This article reviewed the current research progress of neutrophil granules in MI and discusses neutrophil degranulation associated diagnosis and treatment strategies. Video abstract
Collapse
Affiliation(s)
- Nan Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Xiahenazi Aiyasiding
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Wen-Jing Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China
| | - Hai-Han Liao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
57
|
Pasman R, Krom BP, Zaat SAJ, Brul S. The Role of the Oral Immune System in Oropharyngeal Candidiasis-Facilitated Invasion and Dissemination of Staphylococcus aureus. FRONTIERS IN ORAL HEALTH 2022; 3:851786. [PMID: 35464779 PMCID: PMC9021398 DOI: 10.3389/froh.2022.851786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
Candida albicans and Staphylococcus aureus account for most invasive fungal and bacterial bloodstream infections (BSIs), respectively. However, the initial point of invasion responsible for S. aureus BSIs is often unclear. Recently, C. albicans has been proposed to mediate S. aureus invasion of immunocompromised hosts during co-colonization of oral mucosal surfaces. The status of the oral immune system crucially contributes to this process in two distinct ways: firstly, by allowing invasive C. albicans growth during dysfunction of extra-epithelial immunity, and secondly following invasion by some remaining function of intra-epithelial immunity. Immunocompromised individuals at risk of developing invasive oral C. albicans infections could, therefore, also be at risk of contracting concordant S. aureus BSIs. Considering the crucial contribution of both oral immune function and dysfunction, the aim of this review is to provide an overview of relevant aspects of intra and extra-epithelial oral immunity and discuss predominant immune deficiencies expected to facilitate C. albicans induced S. aureus BSIs.
Collapse
Affiliation(s)
- Raymond Pasman
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Bastiaan P. Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sebastian A. J. Zaat
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Stanley Brul
- Department of Molecular Biology and Microbial Food Safety, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Stanley Brul
| |
Collapse
|
58
|
Naqvi I, Giroux N, Olson L, Morrison SA, Llanga T, Akinade TO, Zhu Y, Zhong Y, Bose S, Arvai S, Abramson K, Chen L, Que L, Kraft B, Shen X, Lee J, Leong KW, Nair SK, Sullenger B. DAMPs/PAMPs induce monocytic TLR activation and tolerance in COVID-19 patients; nucleic acid binding scavengers can counteract such TLR agonists. Biomaterials 2022; 283:121393. [PMID: 35349874 PMCID: PMC8797062 DOI: 10.1016/j.biomaterials.2022.121393] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 12/13/2022]
Abstract
Millions of COVID-19 patients have succumbed to respiratory and systemic inflammation. Hyperstimulation of toll-like receptor (TLR) signaling is a key driver of immunopathology following infection by viruses. We found that severely ill COVID-19 patients in the Intensive Care Unit (ICU) display hallmarks of such hyper-stimulation with abundant agonists of nucleic acid-sensing TLRs present in their blood and lungs. These nucleic acid-containing Damage and Pathogen Associated Molecular Patterns (DAMPs/PAMPs) can be depleted using nucleic acid-binding microfibers to limit the patient samples' ability to hyperactivate such innate immune receptors. Single-cell RNA-sequencing revealed that CD16+ monocytes from deceased but not recovered ICU patients exhibit a TLR-tolerant phenotype and a deficient anti-viral response after ex vivo TLR stimulation. Plasma proteomics confirmed such myeloid hyperactivation and revealed DAMP/PAMP carrier consumption in deceased patients. Treatment of these COVID-19 patient samples with MnO nanoparticles effectively neutralizes TLR activation by the abundant nucleic acid-containing DAMPs/PAMPs present in their lungs and blood. Finally, MnO nanoscavenger treatment limits the ability of DAMPs/PAMPs to induce TLR tolerance in monocytes. Thus, treatment with microfiber- or nanoparticle-based DAMP/PAMP scavengers may prove useful for limiting SARS-CoV-2 induced hyperinflammation, preventing monocytic TLR tolerance, and improving outcomes in severely ill COVID-19 patients.
Collapse
Affiliation(s)
- Ibtehaj Naqvi
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | - Nicholas Giroux
- Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA; Duke University, Graduate School, USA
| | - Lyra Olson
- Duke University, Graduate School, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA
| | - Sarah Ahn Morrison
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | | | - Tolu O Akinade
- Columbia University, Department of Biomedical Engineering, USA
| | - Yuefei Zhu
- Columbia University, Department of Biomedical Engineering, USA
| | - Yiling Zhong
- Columbia University, Department of Biomedical Engineering, USA
| | - Shree Bose
- Duke University, Graduate School, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA
| | - Stephanie Arvai
- Duke University Center for Genomic and Computational Biology, RNA Sequencing Core, USA
| | - Karen Abramson
- Duke University Center for Genomic and Computational Biology, RNA Sequencing Core, USA
| | - Lingye Chen
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Loretta Que
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Bryan Kraft
- Duke University School of Medicine, Department of Medicine, Division of Pulmonary Medicine, USA
| | - Xiling Shen
- Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA
| | - Jaewoo Lee
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA
| | - Kam W Leong
- Columbia University, Department of Biomedical Engineering, USA
| | - Smita K Nair
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA; Duke University School of Medicine, Department of Pathology, USA; Duke University School of Medicine, Department of Neurosurgery, USA.
| | - Bruce Sullenger
- Duke University School of Medicine, Department of Surgery, Division of Surgical Sciences, USA; Duke University, Department of Biomedical Engineering, Pratt School of Engineering, USA; Duke University School of Medicine, Department of Pharmacology and Cancer Biology, USA; Duke University School of Medicine, Department of Neurosurgery, USA.
| |
Collapse
|
59
|
Sanches RCO, Mambelli F, Oliveira SC. Neutrophils and schistosomiasis: a missing piece in pathology. Parasite Immunol 2022; 44:e12916. [PMID: 35332932 DOI: 10.1111/pim.12916] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 12/01/2022]
Abstract
Schistosomiasis is a chronic human parasitic disease that causes serious health problems worldwide. The disease-associated liver pathology is one of the hallmarks of infections by S. mansoni and S. japonicum, and is accountable for the debilitating condition found in infected patients. In the past few years, investigative studies have highlighted the key role played by neutrophils and the influence of inflammasome signaling pathway in different pathological conditions. However, it is noteworthy that the study of inflammasome activation in neutrophils has been overlooked by reports concerning macrophages and monocytes. This interplay between neutrophils and inflammasomes is much more poorly investigated during schistosomiasis. Herein we reviewed the role of neutrophils during schistosomiasis and addressed the potential connection between these cells and inflammasome activation in this context.
Collapse
Affiliation(s)
- Rodrigo C O Sanches
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fábio Mambelli
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), CNPq MCT, Salvador, Brazil
| |
Collapse
|
60
|
Simmons SR, Tchalla EYI, Bhalla M, Bou Ghanem EN. The Age-Driven Decline in Neutrophil Function Contributes to the Reduced Efficacy of the Pneumococcal Conjugate Vaccine in Old Hosts. Front Cell Infect Microbiol 2022; 12:849224. [PMID: 35402289 PMCID: PMC8984502 DOI: 10.3389/fcimb.2022.849224] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/28/2022] [Indexed: 01/04/2023] Open
Abstract
Despite the availability of vaccines, Streptococcus pneumoniae (pneumococcus) remains a serious cause of infections in the elderly. The efficacy of anti-pneumococcal vaccines declines with age. While age-driven changes in antibody responses are well defined, less is known about the role of innate immune cells such as polymorphonuclear leukocytes (PMNs) in the reduced vaccine protection seen in aging. Here we explored the role of PMNs in protection against S. pneumoniae in vaccinated hosts. We found that depletion of PMNs in pneumococcal conjugate vaccine (PCV) treated young mice prior to pulmonary challenge with S. pneumoniae resulted in dramatic loss of host protection against infection. Immunization boosted the ability of PMNs to kill S. pneumoniae and this was dependent on bacterial opsonization by antibodies. Bacterial opsonization with immune sera increased several PMN anti-microbial activities including bacterial uptake, degranulation and ROS production. As expected, PCV failed to protect old mice against S. pneumoniae. In probing the role of PMNs in this impaired protection, we found that aging was accompanied by an intrinsic decline in PMN function. PMNs from old mice failed to effectively kill S. pneumoniae even when the bacteria were opsonized with immune sera from young controls. In exploring mechanisms, we found that PMNs from old mice produced less of the antimicrobial peptide CRAMP and failed to efficiently kill engulfed pneumococci. Importantly, adoptive transfer of PMNs from young mice reversed the susceptibility of vaccinated old mice to pneumococcal infection. Overall, this study demonstrates that the age-driven decline in PMN function impairs vaccine-mediated protection against Streptococcus pneumoniae.
Collapse
|
61
|
Mahmud Z, Rahman A, Mishu ID, Kabir Y. Mechanistic insights into the interplays between neutrophils and other immune cells in cancer development and progression. Cancer Metastasis Rev 2022; 41:405-432. [PMID: 35314951 DOI: 10.1007/s10555-022-10024-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/09/2022] [Indexed: 12/12/2022]
Abstract
Cancer is considered a major public health concern worldwide and is characterized by an uncontrolled division of abnormal cells. The human immune system recognizes cancerous cells and induces innate immunity to destroy those cells. However, sustained tumors may protect themselves by developing immune escape mechanisms through multiple soluble and cellular mediators. Neutrophils are the most plenteous leukocytes in the human blood and are crucial for immune defense in infection and inflammation. Besides, neutrophils emancipate the antimicrobial contents, secrete different cytokines or chemokines, and interact with other immune cells to combat and successfully kill cancerous cells. Conversely, many clinical and experimental studies signpost that being a polarized and heterogeneous population with plasticity, neutrophils, particularly their subpopulations, act as a modulator of cancer development by promoting tumor metastasis, angiogenesis, and immunosuppression. Studies also suggest that tumor infiltrating macrophages, neutrophils, and other innate immune cells support tumor growth and survival. Additionally, neutrophils promote tumor cell invasion, migration and intravasation, epithelial to mesenchymal transition, survival of cancer cells in the circulation, seeding, and extravasation of tumor cells, and advanced growth and development of cancer cells to form metastases. In this manuscript, we describe and review recent studies on the mechanisms for neutrophil recruitment, activation, and their interplay with different immune cells to promote their pro-tumorigenic functions. Understanding the detailed mechanisms of neutrophil-tumor cell interactions and the concomitant roles of other immune cells will substantially improve the clinical utility of neutrophils in cancer and eventually may aid in the identification of biomarkers for cancer prognosis and the development of novel therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Atiqur Rahman
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh
| | | | - Yearul Kabir
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
62
|
Stojkov D, Gigon L, Peng S, Lukowski R, Ruth P, Karaulov A, Rizvanov A, Barlev NA, Yousefi S, Simon HU. Physiological and Pathophysiological Roles of Metabolic Pathways for NET Formation and Other Neutrophil Functions. Front Immunol 2022; 13:826515. [PMID: 35251008 PMCID: PMC8889909 DOI: 10.3389/fimmu.2022.826515] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Neutrophils are the most numerous cells in the leukocyte population and essential for innate immunity. To limit their effector functions, neutrophils are able to modulate glycolysis and other cellular metabolic pathways. These metabolic pathways are essential not only for energy usage, but also for specialized effector actions, such as the production of reactive oxygen species (ROS), chemotaxis, phagocytosis, degranulation, and the formation of neutrophil extracellular traps (NETs). It has been demonstrated that activated viable neutrophils can produce NETs, which consists of a DNA scaffold able to bind granule proteins and microorganisms. The formation of NETs requires the availability of increased amounts of adenosine triphosphate (ATP) as it is an active cellular and therefore energy-dependent process. In this article, we discuss the glycolytic and other metabolic routes in association with neutrophil functions focusing on their role for building up NETs in the extracellular space. A better understanding of the requirements of metabolic pathways for neutrophil functions may lead to the discovery of molecular targets suitable to develop novel anti-infectious and/or anti-inflammatory drugs.
Collapse
Affiliation(s)
- Darko Stojkov
- Institute of Pharmacology, University of Bern, Bern, Switzerland.,Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Lea Gigon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shuang Peng
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Robert Lukowski
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Peter Ruth
- Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen, Germany
| | - Alexander Karaulov
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Nickolai A Barlev
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, Russia.,Regulation of Cell Signaling Laboratory, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.,Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia.,Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Institute of Biochemistry, Brandenburg Medical School, Neuruppin, Germany
| |
Collapse
|
63
|
Connelly AN, Huijbregts RPH, Pal HC, Kuznetsova V, Davis MD, Ong KL, Fay CX, Greene ME, Overton ET, Hel Z. Optimization of methods for the accurate characterization of whole blood neutrophils. Sci Rep 2022; 12:3667. [PMID: 35256648 PMCID: PMC8901620 DOI: 10.1038/s41598-022-07455-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/10/2022] [Indexed: 01/25/2023] Open
Abstract
Neutrophils are the most abundant circulating leukocyte population with critical roles in immune defense, regulation of innate and adaptive immune systems, and disease pathogenesis. Our progress in understanding precise mechanisms of neutrophil activation, recruitment, and function has been hampered by the lack of optimized and standardized methods for the characterization and phenotyping of this readily activated population. By comparing eight methods of neutrophil characterization, we demonstrate that the level of neutrophil activation and degranulation is associated with specific experimental conditions and the number and type of manipulation steps employed. Staining whole blood at 4 °C and removal of remaining unbound antibodies prior to one-step fixation and red blood cell lysis minimizes neutrophil activation, decreases phenotypic alterations during processing, and prevents nonspecific antibody binding. The effects of anticoagulants used for collection, processing delays, and time and temperature during sample analysis on neutrophil phenotype are addressed. The presented data provide a foundation for higher quality standards of neutrophil characterization improving consistency and reproducibility among studies.
Collapse
Affiliation(s)
- Ashley N. Connelly
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA ,grid.265892.20000000106344187Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Richard P. H. Huijbregts
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Harish C. Pal
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Valeriya Kuznetsova
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Marcus D. Davis
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Krystle L. Ong
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Christian X. Fay
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Morgan E. Greene
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Edgar T. Overton
- grid.265892.20000000106344187Center for AIDS Research, University of Alabama at Birmingham, Birmingham, AL USA ,grid.265892.20000000106344187Division of Infectious Disease, University of Alabama at Birmingham, Birmingham, AL USA
| | - Zdenek Hel
- grid.265892.20000000106344187Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA ,grid.265892.20000000106344187Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL USA ,grid.265892.20000000106344187Center for AIDS Research, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
64
|
Wu H, Fu M, Xie X, Yang J, Liu Y, Du F, Fang Z, Shang L, Li L. Naples prognostic score, a novel prognostic score for patients with high- and intermediate-risk gastrointestinal stromal tumours after surgical resection. World J Surg Oncol 2022; 20:63. [PMID: 35232450 PMCID: PMC8886834 DOI: 10.1186/s12957-022-02526-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 02/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND A novel multidimensional inflammatory and nutritional assessment system named the Naples prognostic score could serve as an independent prognostic indicator. However, its significance in patients with high- and intermediate-risk gastrointestinal stromal tumours remains unclear. METHODS We performed this retrospective cohort study based on a prospectively collected database of gastrointestinal stromal tumours (GISTs) between March 2010 and December 2019. The Kaplan-Meier method and log-rank test were used for survival analyses. Least absolute shrinkage and selection operator (LASSO) and Cox proportional hazards regression analysis was used for univariate and multivariate analyses. Time-dependent receiver operating characteristic curves were generated to evaluate the discriminatory ability of the prognostic scoring systems. Differences in the areas under the curve were further compared. RESULTS A total of 405 patients with regular follow-up were included and analysed in this study. Significant differences in progression-free survival and overall survival were observed between the groups (P < 0.001). Multivariate analysis demonstrated that the NPS was a significant predictor of poor progression-free survival (1 vs 0, HR = 4.622, P = 0.001; 2 vs 0, HR = 12.770, P < 0.001) and overall survival (2 vs 0, HR = 5.535, P = 0.002). Furthermore, time-dependent AUC analyses showed that the NPS was more accurate than other haematologic prognostic systems. CONCLUSIONS The present study demonstrates that the NPS could independently predict disease progression and survival among patients with high- and intermediate-risk GISTs. The NPS might be regarded and applied as one of the most convenient and effective preoperative risk stratification tools in the future, which should be validated by large-scale multicentre prospective cohort studies.
Collapse
Affiliation(s)
- Hao Wu
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Mengdi Fu
- Department of Clinical Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Xiaozhou Xie
- Department of Gastroenterological Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Jianqiao Yang
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Yang Liu
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Fengying Du
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Zhen Fang
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Liang Shang
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Gastroenterological Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China. .,Department of Digestive Tumor Translational Medicine, Engineering Laboratory of Shandong Province, Shandong Provincial Hospital, Jinan, 250021, Shandong, China. .,Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250021, Shandong, China.
| | - Leping Li
- Department of Gastroenterological Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Gastroenterological Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China. .,Department of Digestive Tumor Translational Medicine, Engineering Laboratory of Shandong Province, Shandong Provincial Hospital, Jinan, 250021, Shandong, China. .,Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250021, Shandong, China.
| |
Collapse
|
65
|
Novel Insights into the Immunomodulatory Effects of Caryophyllane Sesquiterpenes: A Systematic Review of Preclinical Studies. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12052292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Immunomodulation is a key factor in the homeostasis of organisms, both for physiological and inflammatory conditions. In this context, great attention has been devoted to immunomodulant agents, which can boost or modulate the immune system, thus favoring disease relief. The present systematic review is focused on the immunomodulatory properties of plant-based caryophyllane sesquiterpenes, which are unique natural compounds widely studied due to their multiple and pleiotropic bioactivities. Despite lacking clinical evidence, the selected studies highlighted the ability of these substances, especially β-caryophyllene and α-humulene, to modulate the immune system of both in vitro and in vivo models of disease, such as neurodegenerative and inflammatory-based diseases, cancer, and allergies; moreover, some mechanistic hypotheses have been made too. The present overview suggests a further interest in immunomodulation by caryophyllane sesquiterpenes as a possible novel strategy for immune-based diseases or as an adjuvant treatment and encourages further high-quality studies, using high-purity compounds, to better clarify the mechanisms accounting for these properties and to support a further pharmaceutical development.
Collapse
|
66
|
Li W, Cao F, Takase H, Arai K, Lo EH, Lok J. Blood-Brain Barrier Mechanisms in Stroke and Trauma. Handb Exp Pharmacol 2022; 273:267-293. [PMID: 33580391 DOI: 10.1007/164_2020_426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The brain microenvironment is tightly regulated. The blood-brain barrier (BBB), which is composed of cerebral endothelial cells, astrocytes, and pericytes, plays an important role in maintaining the brain homeostasis by regulating the transport of both beneficial and detrimental substances between circulating blood and brain parenchyma. After brain injury and disease, BBB tightness becomes dysregulated, thus leading to inflammation and secondary brain damage. In this chapter, we overview the fundamental mechanisms of BBB damage and repair after stroke and traumatic brain injury (TBI). Understanding these mechanisms may lead to therapeutic opportunities for brain injury.
Collapse
Affiliation(s)
- Wenlu Li
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Fang Cao
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hajime Takase
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Josephine Lok
- Neuroprotection Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
67
|
Hafkamp FM, Mol S, Waqué I, De Jong EC. Dexamethasone, but Not Vitamin D or A, Dampens the Inflammatory Neutrophil Response to Protect At-risk COVID-19 Patients. Immune Netw 2022; 22:e36. [PMID: 36081524 PMCID: PMC9433192 DOI: 10.4110/in.2022.22.e36] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/06/2022] [Accepted: 04/10/2022] [Indexed: 12/15/2022] Open
Abstract
Dexamethasone (DEX) was the first drug shown to save lives of critically ill coronavirus disease 2019 (COVID-19) patients suffering from respiratory distress. A hyperactivated state of neutrophils was found in COVID-19 patients compared to non-COVID pneumonia cases. Given the beneficial effects of DEX in COVID-19 patients, we investigated the effects of DEX and of other immunomodulatory drugs vitamin D3 (VD3) and retinoic acid (RA) on neutrophil function. DEX, but not VD3 or RA, significantly inhibited all tested aspects of neutrophil function, e.g., degranulation, intracellular ROS production, CXCL8 release and NETosis. Interestingly, RA displayed the opposite effect by significantly increasing both CXCL8 and NET release by neutrophils. Taken together, these data suggest that the lower COVID-19 mortality in DEX-treated patients may in part be due to the dampening effect of DEX on the inflammatory neutrophil response, which could prevent neutrophil plugs with NETS in the lungs and other inflamed organs of patients.
Collapse
Affiliation(s)
- Florianne M.J. Hafkamp
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection & Immunity, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Sanne Mol
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection & Immunity, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CS Utrecht, The Netherlands
| | - Iris Waqué
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection & Immunity, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Esther C. De Jong
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection & Immunity, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
68
|
Meier S, Seddon JA, Maasdorp E, Kleynhans L, du Plessis N, Loxton AG, Malherbe ST, Zak DE, Thompson E, Duffy FJ, Kaufmann SHE, Ottenhoff THM, Scriba TJ, Suliman S, Sutherland JS, Winter J, Kuivaniemi H, Walzl G, Tromp G. Neutrophil degranulation, NETosis and platelet degranulation pathway genes are co-induced in whole blood up to six months before tuberculosis diagnosis. PLoS One 2022; 17:e0278295. [PMID: 36454773 PMCID: PMC9714760 DOI: 10.1371/journal.pone.0278295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb) causes tuberculosis (TB) and remains one of the leading causes of mortality due to an infectious pathogen. Host immune responses have been implicated in driving the progression from infection to severe lung disease. We analyzed longitudinal RNA sequencing (RNAseq) data from the whole blood of 74 TB progressors whose samples were grouped into four six-month intervals preceding diagnosis (the GC6-74 study). We additionally analyzed RNAseq data from an independent cohort of 90 TB patients with positron emission tomography-computed tomography (PET-CT) scan results which were used to categorize them into groups with high and low levels of lung damage (the Catalysis TB Biomarker study). These groups were compared to non-TB controls to obtain a complete whole blood transcriptional profile for individuals spanning from early stages of M.tb infection to TB diagnosis. The results revealed a steady increase in the number of genes that were differentially expressed in progressors at time points closer to diagnosis with 278 genes at 13-18 months, 742 at 7-12 months and 5,131 detected 1-6 months before diagnosis and 9,205 detected in TB patients. A total of 2,144 differentially expressed genes were detected when comparing TB patients with high and low levels of lung damage. There was a large overlap in the genes upregulated in progressors 1-6 months before diagnosis (86%) with those in TB patients. A comprehensive pathway analysis revealed a potent activation of neutrophil and platelet mediated defenses including neutrophil and platelet degranulation, and NET formation at both time points. These pathways were also enriched in TB patients with high levels of lung damage compared to those with low. These findings suggest that neutrophils and platelets play a critical role in TB pathogenesis, and provide details of the timing of specific effector mechanisms that may contribute to TB lung pathology.
Collapse
Affiliation(s)
- Stuart Meier
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
| | - James A. Seddon
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
- Desmond Tutu TB Centre, Department of Paediatrics and Child Health, Stellenbosch University, Cape Town, South Africa
- Department of Infectious Diseases, Imperial College London, London, United Kingdom
| | - Elizna Maasdorp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
- Centre for Bioinformatics and Computational Biology, Stellenbosch University, Cape Town, South Africa
| | - Léanie Kleynhans
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Nelita du Plessis
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Andre G. Loxton
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Stephanus T. Malherbe
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Daniel E. Zak
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States of America
| | - Ethan Thompson
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States of America
| | - Fergal J. Duffy
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States of America
| | - Stefan H. E. Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, United States of America
| | - Tom H. M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Sara Suliman
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Jayne S. Sutherland
- Vaccines & Immunity Theme, Medical Research Council Unit, The Gambia, at the London School of Hygiene and Tropical Medicine, Banjul, The Gambia
| | - Jill Winter
- Catalysis Foundation for Health, San Ramon, CA, United States of America
| | - Helena Kuivaniemi
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Gerhard Walzl
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
| | - Gerard Tromp
- Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Stellenbosch University, Cape Town, South Africa
- DSI–NRF Centre of Excellence for Biomedical Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- South African Tuberculosis Bioinformatics Initiative, Stellenbosch University, Cape Town, South Africa
- Centre for Bioinformatics and Computational Biology, Stellenbosch University, Cape Town, South Africa
- * E-mail:
| | | | | |
Collapse
|
69
|
Varghese PM, Mukherjee S, Al-Mohanna FA, Saleh SM, Almajhdi FN, Beirag N, Alkahtani SH, Rajkumari R, Nal Rogier B, Sim RB, Idicula-Thomas S, Madan T, Murugaiah V, Kishore U. Human Properdin Released By Infiltrating Neutrophils Can Modulate Influenza A Virus Infection. Front Immunol 2021; 12:747654. [PMID: 34956182 PMCID: PMC8695448 DOI: 10.3389/fimmu.2021.747654] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The complement system is designed to recognise and eliminate invading pathogens via activation of classical, alternative and lectin pathways. Human properdin stabilises the alternative pathway C3 convertase, resulting in an amplification loop that leads to the formation of C5 convertase, thereby acting as a positive regulator of the alternative pathway. It has been noted that human properdin on its own can operate as a pattern recognition receptor and exert immune functions outside its involvement in complement activation. Properdin can bind directly to microbial targets via DNA, sulfatides and glycosaminoglycans, apoptotic cells, nanoparticles, and well-known viral virulence factors. This study was aimed at investigating the complement-independent role of properdin against Influenza A virus infection. As one of the first immune cells to arrive at the site of IAV infection, we show here that IAV challenged neutrophils released properdin in a time-dependent manner. Properdin was found to directly interact with haemagglutinin, neuraminidase and matrix 1 protein Influenza A virus proteins in ELISA and western blot. Furthermore, modelling studies revealed that properdin could bind HA and NA of the H1N1 subtype with higher affinity compared to that of H3N2 due to the presence of an HA cleavage site in H1N1. In an infection assay using A549 cells, properdin suppressed viral replication in pH1N1 subtype while promoting replication of H3N2 subtype, as revealed by qPCR analysis of M1 transcripts. Properdin treatment triggered an anti-inflammatory response in H1N1-challenged A549 cells and a pro-inflammatory response in H3N2-infected cells, as evident from differential mRNA expression of TNF-α, NF-κB, IFN-α, IFN-β, IL-6, IL-12 and RANTES. Properdin treatment also reduced luciferase reporter activity in MDCK cells transduced with H1N1 pseudotyped lentiviral particles; however, it was increased in the case of pseudotyped H3N2 particles. Collectively, we conclude that infiltrating neutrophils at the site of IAV infection can release properdin, which then acts as an entry inhibitor for pandemic H1N1 subtype while suppressing viral replication and inducing an anti-inflammatory response. H3N2 subtype can escape this immune restriction due to altered haemagglutinin and neuraminindase, leading to enhanced viral entry, replication and pro-inflammatory response. Thus, depending on the subtype, properdin can either limit or aggravate IAV infection in the host.
Collapse
Affiliation(s)
- Praveen M Varghese
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom.,School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Shuvechha Mukherjee
- Biomedical Informatics Centre, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Futwan A Al-Mohanna
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Souad M Saleh
- Department of Cell Biology, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Fahad N Almajhdi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nazar Beirag
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Saad H Alkahtani
- Department of Zoology, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Reena Rajkumari
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Beatrice Nal Rogier
- INSERM U1104 Centre d'immunologie de Marseille-Luminy (CIML), Marseille, France
| | - Robert B Sim
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Susan Idicula-Thomas
- Biomedical Informatics Centre, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Taruna Madan
- Department of Innate Immunity, Indian Council of Medical Research (ICMR)-National Institute for Research in Reproductive Health, Mumbai, India
| | - Valarmathy Murugaiah
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Uday Kishore
- Biosciences, College of Health, Medicine and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
70
|
Exploration of the Key Proteins of High-Grade Intraepithelial Neoplasia to Adenocarcinoma Sequence Using In-Depth Quantitative Proteomics Analysis. JOURNAL OF ONCOLOGY 2021; 2021:5538756. [PMID: 34880916 PMCID: PMC8648452 DOI: 10.1155/2021/5538756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 11/09/2021] [Indexed: 11/26/2022]
Abstract
Purpose In this study, we aimed to provide a comprehensive description of typical features and identify key proteins associated with the high-grade intraepithelial neoplasia- (HIN-) adenocarcinoma (AC) sequence. Methods We conducted tandem mass tag-based quantitative proteomic profiling of normal mucosa, HIN, and AC tissues. Protein clusters representative of the HIN-AC sequence were identified using heatmaps based on Pearson's correlation analysis. Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Reactome analyses were performed using the Database for Annotation, Visualization, and Integrated Discovery (DAVID) database, ClueGO plugin in Cytoscape, and the Metascape database. The prognostic value of the key proteins and their effects on the tumor microenvironment and consensus molecular subtype were explored based on The Cancer Genome Atlas. Results We identified 536 proteins categorized into three clusters. Among the biological processes and pathways of the highly expressed proteins in the HIN-AC sequence, proteins were predominantly enriched in response to gut microbiota, cell proliferation, leukocyte migration, and extracellular matrix (ECM) organization events. SERPINH1 and P3H1 were identified as the key proteins that promote the HIN-AC sequence. In the correlation analysis of infiltrating immune cells, both SERPINH1 and P3H1 expression correlated negatively with tumor purity, while correlating positively with abundance of CD8+ T cells, B cells, macrophage/monocytes, dendritic cells, cancer-associated fibroblasts, endothelial cells, neutrophils, and natural killer cells. Furthermore, both SERPINH1 and P3H1 expression positively correlated with common immune checkpoints and mesenchymal molecular subtype. High P3H1 expression was associated with poor disease-free survival and overall survival. Conclusions ECM-related biological processes and pathways are typical features of the HIN-AC sequence. SERPINH1 and P3H1 might be the key proteins in this sequence and be related to ECM remodeling and immune suppression status in CRC.
Collapse
|
71
|
Samukawa S, Yoshimi R, Kirino Y, Nakajima H. The PRY/SPRY domain of pyrin/TRIM20 interacts with β 2-microglobulin to promote inflammasome formation. Sci Rep 2021; 11:23613. [PMID: 34880353 PMCID: PMC8654936 DOI: 10.1038/s41598-021-03073-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 11/26/2021] [Indexed: 01/04/2023] Open
Abstract
Pyrin/TRIM20 is expressed in the neutrophils and monocytes/macrophages and regulates caspase-1 activation and interleukin-1β maturation. Although the mutations in the PRY/SPRY domain of pyrin cause familial Mediterranean fever (FMF), the mechanism of how mutated pyrin provokes excessive inflammation in FMF patients is not well understood. The present study investigated the role of pyrin/TRIM20 in inflammation and the pathogenesis of FMF. β2-Microglobulin (β2MG) was identified as the novel pyrin ligand binding to the PRY/SPRY domain by yeast two-hybrid screenings and co-immunoprecipitation analysis. β2MG was co-localized with pyrin not only in the HEK293 cells overexpressing these proteins but also in the monosodium urate-stimulated human neutrophils in the speck-like structures. The pyrin–β2MG interaction triggered the binding of pyrin and proline–serine–threonine phosphatase interacting protein 1 (PSTPIP1) and then the subsequent recruitment of apoptosis-associated speck-like protein containing caspase recruitment domain (ASC). Caspase-1 p20 subunit, produced by pyrin inflammasome, also interacted with the pyrin PRY/SPRY domain and inhibited the pyrin–β2MG interaction. FMF-associated pyrin mutation M694V did not affect pyrin–β2MG interaction but weakened this inhibition. Our findings suggest that β2MG functions as the pyrin ligand inducing pyrin inflammasome formation and that the FMF-associated pyrin mutations weakened negative feedback of caspase-1 p20 subunit.
Collapse
Affiliation(s)
- Sei Samukawa
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Ryusuke Yoshimi
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
| | - Yohei Kirino
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Hideaki Nakajima
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| |
Collapse
|
72
|
The Effect of Curcuma longa on Inflammatory Mediators and Immunological, Oxidant, and Antioxidant Biomarkers in Asthmatic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4234326. [PMID: 34804178 PMCID: PMC8604592 DOI: 10.1155/2021/4234326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 12/26/2022]
Abstract
The effects of Curcuma longa (C. longa) on total and differential WBC, inflammatory and immunologic mediators, and oxidant and antioxidant biomarkers in bronchoalveolar lavage fluid (BALF) of rats model of asthma were assessed. Animals were divided to 5 groups including control (C), asthma (sensitized to ovalbumin), and asthmatic groups treated with 0.75, 1.50, and 3.00 mg/ml C. longa (CL) and 1.25 μg/ml dexamethasone (D) (8 rats in each group). Total and differential WBC count, concentrations of phospholipase A2 (PLA2), total protein (TP), interferon-gamma (IFN-γ), interleukin-4 (IL-4), immunoglobulin E (IgE), NO2, NO3, malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), and thiol in BALF were assessed. Total and most differential WBC counts and BALF levels of PLA2, TP, IgE, IL-4, and oxidants in asthma group were higher but antioxidants and IFN-γ levels as well as IFN-γ/IL-4 ratio were lower than control group (p < 0.001 for all cases). Total WBC and levels of PLA2, IgE, NO2, and NO3 were significantly reduced following treatment with C. longa, compared to asthma group (p < 0.001 for all cases). In groups treated with dexamethasone and two higher concentrations of C. longa, neutrophil and eosinophil counts as well as TP, IL-4, and MDA levels were significantly decreased but IFN-γ, IFN-γ/IL-4 ratio, and antioxidants were increased (except IFN-γ/IL-4 ratio), compared to asthma group (p < 0.05 to p < 0.001). Compared to dexamethasone, C. longa exerted more pronounced effects on lung inflammation, oxidative stress, and immune system in asthmatic rats.
Collapse
|
73
|
Subhan MA, Torchilin VP. Neutrophils as an emerging therapeutic target and tool for cancer therapy. Life Sci 2021; 285:119952. [PMID: 34520766 DOI: 10.1016/j.lfs.2021.119952] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 02/09/2023]
Abstract
Activation of neutrophils is necessary for the protection of the host against microbial infection. This property can be used as mode of therapy for cancer treatment. Neutrophils have conflicting dual functions in cancer as either a tumor promoter or inhibitor. Neutrophil-based drug delivery has achieved increased attention in pre-clinical models. This review addresses in detail the different neutrophil constituents, the conflicting function of neutrophils and activation of the neutrophil as an important target of therapy for cancer treatment, and use of neutrophils or neutrophil membrane-derived vesicles as vehicles for drug delivery and targeting.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh..
| | - Vladimir P Torchilin
- CPBN, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA; Department of Oncology, Radiotherapy and Plastic Surgery, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia.
| |
Collapse
|
74
|
Chen H, Wu X, Xu C, Lin J, Liu Z. Dichotomous roles of neutrophils in modulating pathogenic and repair processes of inflammatory bowel diseases. PRECISION CLINICAL MEDICINE 2021; 4:246-257. [PMID: 35692862 PMCID: PMC8982532 DOI: 10.1093/pcmedi/pbab025] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023] Open
Abstract
Neutrophils are considered as complex innate immune cells and play a critical role in maintaining intestinal mucosal homeostasis. They exert robust pro-inflammatory effects and recruit other immune cells in the acute phase of pathogen infection and intestinal inflammation, but paradoxically, they also limit exogenous microbial invasion and facilitate mucosal restoration. Hyperactivation or dysfunction of neutrophils results in abnormal immune responses, leading to multiple autoimmune and inflammatory diseases including systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel diseases (IBD). As a refractory intestinal inflammatory disease, the pathogenesis and progression of IBD are associated with complicated immune response processes in which neutrophils are profoundly involved. However, the consensus on potential roles of neutrophils in modulating pathogenic and repair processes of IBD remains not fully understood. Accumulated infiltrating neutrophils cross the epithelial barrier and contribute to microbial dysbiosis, aggravated intestinal architectural damage, compromised resolution of intestinal inflammation and increased risk of thrombosis during IBD. Paradoxically, activated neutrophils are also associated with effective elimination of invaded microbiota, promoted angiogenesis and tissue restoration of gut mucosa in IBD. Here, we discuss the beneficial and detrimental roles of neutrophils in the onset and resolution of intestinal mucosal inflammation, hoping to provide a precise overview of neutrophil functions in the pathogenesis of IBD.
Collapse
Affiliation(s)
- Huimin Chen
- Center for Inflammatory Bowel Disease Research, the Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiaohan Wu
- Center for Inflammatory Bowel Disease Research, the Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Chunjin Xu
- Department of Gastroenterology, the First People's Hospital of Shangqiu City Affiliated to Xinxiang Medical University, Shangqiu 476100, China
| | - Jian Lin
- Department of Gastroenterology, Affiliated Hospital of Putian University, Putian 351106, China
| | - Zhanju Liu
- Center for Inflammatory Bowel Disease Research, the Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
75
|
Oliveira-Costa KM, Menezes GB, Paula Neto HA. Neutrophil accumulation within tissues: A damage x healing dichotomy. Biomed Pharmacother 2021; 145:112422. [PMID: 34781139 DOI: 10.1016/j.biopha.2021.112422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 02/09/2023] Open
Abstract
The abundance of neutrophils in human circulation, their fast mobilization from blood to tissues, along with their alleged short life-span led to the image of neutrophils as a homogeneous cell type designed to fight infections and die in the process. Additionally, their granule content and capacity to produce molecules with considerable cytotoxic potential, lead to the general belief that neutrophil activation inexorably results in side effect of extensive tissue injury. Neutrophil activation in fact causes tissue injury as an adverse effect, but it seems that this is restricted to particular pathological situations and more of an "exception to the rule". Here we review evidences arising especially from intravital microscopy studies that demonstrate neutrophils as cells endowed with sophisticated mechanisms and able to engage in complex interactions as to minimize damage and optimize their effector functions. Moreover, neutrophil infiltration may even contribute to tissue healing and repair which may altogether demand a reexamination of current anti-inflammatory therapies that have neutrophil migration and activation as a target.
Collapse
Affiliation(s)
- Karen Marques Oliveira-Costa
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil
| | - Gustavo B Menezes
- Center for Gastrointestinal Biology, Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais 31270-901, Brazil.
| | - Heitor A Paula Neto
- Laboratório de Alvos Moleculares, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
76
|
Probiotics in Counteracting the Role of Neutrophils in Cancer Metastasis. Vaccines (Basel) 2021; 9:vaccines9111306. [PMID: 34835236 PMCID: PMC8621509 DOI: 10.3390/vaccines9111306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 12/20/2022] Open
Abstract
Neutrophils are known for their role geared towards pathogen clearance by different mechanisms that they initiate, primarily by the release of neutrophil extracellular traps (NETs). However, their immune-surveillance capacity accompanied with plasticity in existing as interchangeable subsets, discovered recently, has revealed their property to contribute to complex cancer pathologies including tumor initiation, growth, angiogenesis and metastasis. Although there is a growing body of evidence suggesting a critical balance between the protumoral and antitumoral neutrophil phenotypes, an in-depth signaling pathway analysis would aid in determination of anticipatory, diagnostic and therapeutic targets. This review presents a comprehensive overview of the potential pathways involved in neutrophil-triggered cancer metastasis and introduces the influence of the microbial load and avenues for probiotic intervention.
Collapse
|
77
|
Abstract
A considerable amount of continuous proliferation and differentiation is required to produce daily a billion new neutrophils in an adult human. Of the few cytokines and factors known to control neutrophil production, G-CSF is the guardian of granulopoiesis. G-CSF/CSF3R signaling involves the recruitment of non-receptor protein tyrosine kinases and their dependent signaling pathways of serine/threonine kinases, tyrosine phosphatases, and lipid second messengers. These pathways converge to activate the families of STAT and C/EBP transcription factors. CSF3R mutations are associated with human disorders of neutrophil production, including severe congenital neutropenia, neutrophilia, and myeloid malignancies. More than three decades after their identification, cloning, and characterization of G-CSF and G-CSF receptor, fundamental questions remain about their physiology.
Collapse
Affiliation(s)
- Hrishikesh M Mehta
- Departments of Cancer Biology and Pediatrics, Lerner Research Institute at the Cleveland Clinic, United States
| | - Seth J Corey
- Departments of Cancer Biology and Pediatrics, Lerner Research Institute at the Cleveland Clinic, United States.
| |
Collapse
|
78
|
Apoptotic cell-derived metabolites in efferocytosis-mediated resolution of inflammation. Cytokine Growth Factor Rev 2021; 62:42-53. [PMID: 34742632 DOI: 10.1016/j.cytogfr.2021.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 10/13/2021] [Indexed: 12/21/2022]
Abstract
The resolution of inflammation, as part of standard host defense mechanism, is the process to guarantee timely termination of inflammatory responses and eventual restoration of tissue homeostasis . It is mainly achieved via efferocytosis, during which pro-resolving macrophages clear apoptotic neutrophils at the inflammatory site. Unfortunately, impaired resolution can be the leading cause of chronic inflammatory disorders and some autoimmune diseases. Existing studies have provided relatively comprehensive understandings about the recognition and uptake of apoptotic neutrophils by macrophages during early phases of efferocytosis. However, lack of information concerns macrophage metabolism of apoptotic cell-derived metabolites after being released from phagolysosomes or the relationship between such metabolism and efferocytosis. Notwithstanding, three recent studies have revealed macrophage metabolism of cholesterol, fatty acids and arginine, as well as their respective functions in the context of inflammation-resolution. This review provides an overview of the resolution of inflammation, efferocytosis and the key players involved, followed by a focus on the metabolism of apoptotic cell-derived metabolites within efferocytes. Hypotheses of more potential apoptotic cell-derived metabolites and their possible roles in the resolution are also formulated. Understanding the effect of these metabolites further advances the concept that apoptotic cells act as active players to regulate resolution, and also suggests novel therapeutic strategies for diseases driven by defective resolution and even cancer that may be treated through enhanced efferocytosis.
Collapse
|
79
|
Amanso AM, Turner TC, Kamalakar A, Ballestas SA, Hymel LA, Randall J, Johnston R, Arthur RA, Willett NJ, Botchwey EA, Goudy SL. Local delivery of FTY720 induces neutrophil activation through chemokine signaling in an oronasal fistula model. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021; 7:160-174. [PMID: 34722855 PMCID: PMC8549964 DOI: 10.1007/s40883-021-00208-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 03/07/2021] [Accepted: 03/15/2021] [Indexed: 11/07/2022]
Abstract
Purpose Cleft palate repair surgeries lack a regenerative reconstructive option and, in many cases, develop complications including oronasal fistula (ONF). Our group has developed a novel murine phenocopy of ONF to study the oral cavity wound healing program. Using this model, our team previously identified that delivery of FTY720 on a nanofiber scaffold had a unique immunomodulatory effect directing macrophages and monocytes into a pro-regenerative state during ONF healing. Here, the objective of this study was to determine the effects of local biomaterial-based FTY720 delivery in the ONF model on the early bulk gene expression and neutrophil phenotypic response within the regenerating tissue. Methods Using a mouse model of ONF formation, a palate defect was created and was treated with FTY720 nanofiber scaffolds or (blank) vehicle control nanofibers. At 1 and 3 days post-implantation, ONF oral mucosal tissue from the defect region was collected for RNA sequencing analysis or flow cytometry. For the RNA-seq expression profiling, intracellular pathways were assessed using the KEGG Pathway database and Gene Ontology (GO) Terms enrichment interactive graph. To assess the effects of FTY720 on different neutrophil subpopulations, flow cytometry data was analyzed using pseudotime analysis based on Spanning-tree Progression Analysis of Density-normalized Events (SPADE). Results RNA sequencing analysis of palate mucosa injured tissue identified 669 genes that were differentially expressed (DE) during the first 3 days of ONF wound healing after local delivery of FTY720, including multiple genes in the sphingolipid signaling pathway. Evaluation of the DE genes at the KEGG Pathway database also identified the inflammatory immune response pathways (chemokine signaling, cytokine-cytokine receptor interaction, and leukocyte transendothelial migration), and the Gene Ontology enrichment analysis identified neutrophil chemotaxis and migration terms. SPADE dendrograms of CD11b+Ly6G+ neutrophils at both day 1 and day 3 post-injury showed significantly distinct subpopulations of neutrophils in oral mucosal defect tissue from the FTY720 scaffold treatment group compared to the vehicle control group (blank). Increased expression of CD88 and Vav1, among other genes, were found and staining of the ONF area demonstrated increased VAV1 staining in FTY720‐treated healing oral mucosa. Conclusion Treatment of oral mucosal defects using FTY720 scaffolds is a promising new immunotherapy to improve healing outcomes and reducing ONF formation during cleft palate surgical repair. Local delivery of FTY720 nanofiber scaffolds during ONF healing significantly shifted early gene transcription associated with immune cell recruitment and modulation of the immune microenvironment results in distinct neutrophil subpopulations in the oral mucosal defect tissue that provides a critical shift toward pro-regenerative immune signaling. Supplementary Information The online version contains supplementary material available at 10.1007/s40883-021-00208-z.
Collapse
Affiliation(s)
- A M Amanso
- Department of Otolaryngology, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322 USA
| | - T C Turner
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA.,Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA
| | - A Kamalakar
- Department of Otolaryngology, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322 USA
| | - S A Ballestas
- Department of Otolaryngology, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322 USA
| | - L A Hymel
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA.,Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA
| | - J Randall
- The Emory Integrated Computational Core, Emory University School of Medicine, Atlanta, GA USA
| | - R Johnston
- The Emory Integrated Computational Core, Emory University School of Medicine, Atlanta, GA USA
| | - R A Arthur
- The Emory Integrated Computational Core, Emory University School of Medicine, Atlanta, GA USA
| | - N J Willett
- Department of Orthopedics, Emory University School of Medicine, Atlanta, GA USA.,Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA.,Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA
| | - E A Botchwey
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA.,Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA USA
| | - S L Goudy
- Department of Otolaryngology, Emory University School of Medicine, 2015 Uppergate Drive, Atlanta, GA 30322 USA
| |
Collapse
|
80
|
Volpedo G, Pacheco-Fernandez T, Bhattacharya P, Oljuskin T, Dey R, Gannavaram S, Satoskar AR, Nakhasi HL. Determinants of Innate Immunity in Visceral Leishmaniasis and Their Implication in Vaccine Development. Front Immunol 2021; 12:748325. [PMID: 34712235 PMCID: PMC8546207 DOI: 10.3389/fimmu.2021.748325] [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: 07/27/2021] [Accepted: 09/24/2021] [Indexed: 12/22/2022] Open
Abstract
Leishmaniasis is endemic to the tropical and subtropical regions of the world and is transmitted by the bite of an infected sand fly. The multifaceted interactions between Leishmania, the host innate immune cells, and the adaptive immunity determine the severity of pathogenesis and disease development. Leishmania parasites establish a chronic infection by subversion and attenuation of the microbicidal functions of phagocytic innate immune cells such as neutrophils, macrophages and dendritic cells (DCs). Other innate cells such as inflammatory monocytes, mast cells and NK cells, also contribute to resistance and/or susceptibility to Leishmania infection. In addition to the cytokine/chemokine signals from the innate immune cells, recent studies identified the subtle shifts in the metabolic pathways of the innate cells that activate distinct immune signal cascades. The nexus between metabolic pathways, epigenetic reprogramming and the immune signaling cascades that drive the divergent innate immune responses, remains to be fully understood in Leishmania pathogenesis. Further, development of safe and efficacious vaccines against Leishmaniasis requires a broader understanding of the early interactions between the parasites and innate immune cells. In this review we focus on the current understanding of the specific role of innate immune cells, the metabolomic and epigenetic reprogramming and immune regulation that occurs during visceral leishmaniasis, and the strategies used by the parasite to evade and modulate host immunity. We highlight how such pathways could be exploited in the development of safe and efficacious Leishmania vaccines.
Collapse
Affiliation(s)
- Greta Volpedo
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Thalia Pacheco-Fernandez
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Parna Bhattacharya
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Timur Oljuskin
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Ranadhir Dey
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Sreenivas Gannavaram
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| | - Abhay R Satoskar
- Departments of Pathology and Microbiology, Wexner Medical Center, The Ohio State University, Columbus, OH, United States
| | - Hira L Nakhasi
- Laboratory of Emerging Pathogens, Division of Emerging and Transfusion Transmitted Diseases, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
81
|
Peng Z, Zhao C, Du X, Yang Y, Li Y, Song Y, Fang B, Zhang Y, Qin X, Zhang Y, Li X, Wang Z, Li X, Liu G. Autophagy Induced by Palmitic Acid Regulates Neutrophil Adhesion Through the Granule-Dependent Degradation of αMβ2 Integrin in Dairy Cows With Fatty Liver. Front Immunol 2021; 12:726829. [PMID: 34691032 PMCID: PMC8529007 DOI: 10.3389/fimmu.2021.726829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/02/2021] [Indexed: 11/28/2022] Open
Abstract
β2 integrins are critical for neutrophil firm adhesion, trans-endothelial migration, and the recruitment to the inflamed tissue. Autophagy is implicated in cell migration and tumor metastasis through facilitating the turnover of β1 integrins; however, whether autophagy is able to control neutrophil migration by promoting the degradation of β2 integrins is unexplored. Here, we show that high blood levels of palmitic acid (PA) strongly triggered neutrophil autophagy activation, leading to adhesion deficiency in dairy cows with fatty liver. The three neutrophil granule subtypes, namely, azurophil granules (AGs), specific granules (SGs), and gelatinase granules (GGs), were engulfed by the autophagosomes for degradation, resulting in an increased vacuolation in fatty liver dairy cow neutrophils. Importantly, the adhesion-associated molecules CD11b and CD18 distributed on AGs, SGs, and GGs were degraded with the three granule subtypes by autophagy. Moreover, FGA, Hsc70, and TRIM21 mediated the degradation of cytosolic oxidized–ubiquitinated CD11b and CD18. Collectively, our results demonstrate that high blood PA triggers neutrophil autophagy-dependent vacuolation and granule-dependent adhesion deficiency, decreasing neutrophil mobility, and impairing the innate immune system of dairy cow with fatty liver. This theory extends the category of autophagy in maintaining granule homeostasis and provides a novel strategy to improve the immune of dairy cows with metabolic disease.
Collapse
Affiliation(s)
- Zhicheng Peng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chenxu Zhao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuchen Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yunfei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Baochen Fang
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Yuming Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xia Qin
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuanyuan Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaobing Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
82
|
Zhu S, Yu Y, Ren Y, Xu L, Wang H, Ling X, Jin L, Hu Y, Zhang H, Miao C, Guo K. The emerging roles of neutrophil extracellular traps in wound healing. Cell Death Dis 2021; 12:984. [PMID: 34686654 PMCID: PMC8536667 DOI: 10.1038/s41419-021-04294-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/27/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022]
Abstract
Delayed wound healing causes problems for many patients both physically and psychologically, contributing to pain, economic burden, loss of function, and even amputation. Although many factors affect the wound healing process, abnormally prolonged or augmented inflammation in the wound site is a common cause of poor wound healing. Excessive neutrophil extracellular trap (NET) formation during this phase may amplify inflammation and hinder wound healing. However, the roles of NETs in wound healing are still unclear. Herein, we briefly introduce NET formation and discuss the possible NET-related mechanisms in wound healing. We conclude with a discussion of current studies, focusing on the roles of NETs in diabetic and normoglycemic wounds and the effectiveness of NET-targeting treatments in wound healing.
Collapse
Affiliation(s)
- Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Ren
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liying Xu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huilin Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaomin Ling
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Jin
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
83
|
Combadière B, Adam L, Guillou N, Quentric P, Rosenbaum P, Dorgham K, Bonduelle O, Parizot C, Sauce D, Mayaux J, Luyt CE, Boissonnas A, Amoura Z, Pourcher V, Miyara M, Gorochov G, Guihot A, Combadière C. LOX-1-Expressing Immature Neutrophils Identify Critically-Ill COVID-19 Patients at Risk of Thrombotic Complications. Front Immunol 2021; 12:752612. [PMID: 34616409 PMCID: PMC8488276 DOI: 10.3389/fimmu.2021.752612] [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] [Received: 08/04/2021] [Accepted: 09/07/2021] [Indexed: 12/23/2022] Open
Abstract
Background Lymphopenia and the neutrophil/lymphocyte ratio may have prognostic value in COVID-19 severity. Objective We investigated neutrophil subsets and functions in blood and bronchoalveolar lavage (BAL) of COVID-19 patients on the basis of patients’ clinical characteristics. Methods We used a multiparametric cytometry profiling based to mature and immature neutrophil markers in 146 critical or severe COVID-19 patients. Results The Discovery study (38 patients, first pandemic wave) showed that 80% of Intensive Care Unit (ICU) patients develop strong myelemia with CD10−CD64+ immature neutrophils (ImNs). Cellular profiling revealed three distinct neutrophil subsets expressing either the lectin‐like oxidized low‐density lipoprotein receptor‐1 (LOX‐1), the interleukin-3 receptor alpha (CD123), or programmed death-ligand 1 (PD-L1) overrepresented in ICU patients compared to non-ICU patients. The proportion of LOX-1- or CD123-expressing ImNs is positively correlated with clinical severity, cytokine storm (IL-1β, IL-6, IL-8, TNFα), acute respiratory distress syndrome (ARDS), and thrombosis. BALs of patients with ARDS were highly enriched in LOX-1-expressing ImN subsets and in antimicrobial neutrophil factors. A validation study (118 patients, second pandemic wave) confirmed and strengthened the association of the proportion of ImN subsets with disease severity, invasive ventilation, and death. Only high proportions of LOX-1-expressing ImNs remained strongly associated with a high risk of severe thrombosis independently of the plasma antimicrobial neutrophil factors, suggesting an independent association of ImN markers with their functions. Conclusion LOX-1-expressing ImNs may help identifying COVID-19 patients at high risk of severity and thrombosis complications.
Collapse
Affiliation(s)
- Behazine Combadière
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Lucille Adam
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Noëlline Guillou
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Paul Quentric
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Pierre Rosenbaum
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Karim Dorgham
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Olivia Bonduelle
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Christophe Parizot
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Delphine Sauce
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Julien Mayaux
- Assistance Publique - Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Service de Pneumologie, Médecine Intensive et Réanimation, Paris, France
| | - Charles-Edouard Luyt
- Service de Médecine Intensive Réanimation, Institut de Cardiologie, Assistance Publique - Hôpitaux de Paris (AP-HP), Sorbonne Université, Hôpital Pitié - Salpêtrière, Paris, France.,Sorbonne Université, Inserm, Institute of Cardiometabolism and Nutrition (ICAN), Paris, France
| | - Alexandre Boissonnas
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| | - Zahir Amoura
- Service de Médecine Interne 2, Institut E3M, Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Valérie Pourcher
- Assistance Publique - Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Service de Maladies infectieuses et Tropicales, Paris, France
| | - Makoto Miyara
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Guy Gorochov
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Amélie Guihot
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France.,Assistance Publique - Hôpitaux de Paris (AP-HP), Groupement Hospitalier Pitié-Salpêtrière, Département d'Immunologie, Paris, France
| | - Christophe Combadière
- Sorbonne Université, Institut national de santé et de recherche medicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Centre d'Immunologie et des Maladies Infectieuses, Cimi-Paris, Paris, France
| |
Collapse
|
84
|
Fahed AC, Jang IK. Plaque erosion and acute coronary syndromes: phenotype, molecular characteristics and future directions. Nat Rev Cardiol 2021; 18:724-734. [PMID: 33953381 DOI: 10.1038/s41569-021-00542-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2021] [Indexed: 02/03/2023]
Abstract
Although acute coronary syndromes (ACS) remain one of the leading causes of death, the clinical presentation has changed over the past three decades with a decline in the incidence of ST-segment elevation myocardial infarction (STEMI) and an increase in non-STEMI. This epidemiological shift is at least partially explained by changes in plaque biology as a result of the widespread use of statins. Historically, atherosclerotic plaque rupture of the fibrous cap was thought to be the main culprit in ACS. However, plaque erosion with an intact fibrous cap is now responsible for about one third of ACS and up to two thirds of non-STEMI. Two major research approaches have enabled a better understanding of plaque erosion. First, advanced intravascular imaging has provided opportunities for an 'optical biopsy' and extensive phenotyping of coronary plaques in living patients. Second, basic science experiments have shed light on the unique molecular characteristics of plaque erosion. At present, patients with ACS are still uniformly treated with coronary stents irrespective of the underlying pathobiology. However, pilot studies indicate that patients with plaque erosion might be treated conservatively without coronary stenting. In this Review, we discuss the patient phenotype and the molecular characteristics in atherosclerotic plaque erosion and provide our vision for a potential major shift in the management of patients with plaque erosion.
Collapse
Affiliation(s)
- Akl C Fahed
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ik-Kyung Jang
- Cardiology Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Kyung Hee University, Seoul, South Korea.
| |
Collapse
|
85
|
Mol S, Hafkamp FMJ, Varela L, Simkhada N, Taanman-Kueter EW, Tas SW, Wauben MHM, Groot Kormelink T, de Jong EC. Efficient Neutrophil Activation Requires Two Simultaneous Activating Stimuli. Int J Mol Sci 2021; 22:ijms221810106. [PMID: 34576270 PMCID: PMC8467451 DOI: 10.3390/ijms221810106] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are abundantly present in the synovium and synovial fluid of patients suffering from arthritis. Neutrophils can be activated by a multitude of stimuli and the current dogma states that this is a two-step process, consisting of a priming step followed by an activation step. Considering that neutrophil activation occurs in an inflammatory environment, where multiple stimuli are present, we argue that a two-step process is highly unlikely. Here, we indeed demonstrate that neutrophils require simultaneous ligation of two different receptors for efficient activation. We isolated human peripheral blood neutrophils and cultured them with various combinations of stimuli (GM-CSF, fMLF, TNF, and LPS). Next, we evaluated essential neutrophil functions, including degranulation and ROS production using flow cytometry, mediator release using ELISA, NETosis by a live cell imaging method, phagocytosis by imaging flow cytometry, and extracellular vesicle (EV) release quantified by high-resolution flow cytometry. Exposure of neutrophils to any combination of stimuli, but not to single stimuli, resulted in significant degranulation, and mediator and EV release. Furthermore, ROS production increased substantially by dual stimulation, yet appeared to be more dependent on the type of stimulation than on dual stimulation. Phagocytosis was induced to its maximum capacity by a single stimulus, while NETosis was not induced by any of the used physiological stimuli. Our data indicate that neutrophil activation is tightly regulated and requires activation by two simultaneous stimuli, which is largely independent of the combination of stimuli.
Collapse
Affiliation(s)
- Sanne Mol
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
- Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, 3508 TD Utrecht, The Netherlands; (L.V.); (M.H.M.W.)
| | - Florianne M. J. Hafkamp
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
| | - Laura Varela
- Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, 3508 TD Utrecht, The Netherlands; (L.V.); (M.H.M.W.)
| | - Neena Simkhada
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
| | - Esther W. Taanman-Kueter
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
| | - Sander W. Tas
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
- Amsterdam Rheumatology and Immunology Center, Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Marca H. M. Wauben
- Department Biomolecular Health Sciences, Faculty Veterinary Medicine, Utrecht University, 3508 TD Utrecht, The Netherlands; (L.V.); (M.H.M.W.)
| | - Tom Groot Kormelink
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
| | - Esther C. de Jong
- Department Experimental Immunology, Amsterdam UMC, Location AMC, 1105 AZ Amsterdam, The Netherlands; (S.M.); (F.M.J.H.); (N.S.); (E.W.T.-K.); (S.W.T.); (T.G.K.)
- Correspondence: ; Tel.: +31-2-0566-4963
| |
Collapse
|
86
|
|
87
|
Inhibition of elastase enhances the adjuvanticity of alum and promotes anti-SARS-CoV-2 systemic and mucosal immunity. Proc Natl Acad Sci U S A 2021; 118:2102435118. [PMID: 34353890 PMCID: PMC8403952 DOI: 10.1073/pnas.2102435118] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
We report that suppression of the serine protease elastase reshapes innate responses induced by injected vaccines containing alum adjuvant. This reprogramming improves the induction of protective antibodies in the bloodstream and stimulates innate signals, which support the development of antibody responses in mucosal tissues. Our findings identify elastase as the innate regulator that blunts the adjuvant activity of alum. They also demonstrate that vaccination via mucosal routes is not an absolute requirement for antibody responses in mucosal tissues and secretions. Supplementation of an alum-based vaccine containing SARS-CoV-2 spike protein subunit 1 as antigen increased anti–SARS-CoV-2 immunity in the blood and mucosal secretions in mice. Thus, this strategy could help in the development of future protein-based vaccines against SARS-CoV-2. Alum, used as an adjuvant in injected vaccines, promotes T helper 2 (Th2) and serum antibody (Ab) responses. However, it fails to induce secretory immunoglobulin (Ig) A (SIgA) in mucosal tissues and is poor in inducing Th1 and cell-mediated immunity. Alum stimulates interleukin 1 (IL-1) and the recruitment of myeloid cells, including neutrophils. We investigated whether neutrophil elastase regulates the adjuvanticity of alum, and whether a strategy targeting neutrophil elastase could improve responses to injected vaccines. Mice coadministered a pharmacological inhibitor of elastase, or lacking elastase, developed high-affinity serum IgG and IgA antibodies after immunization with alum-adsorbed protein vaccines, including the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2). These mice also developed broader antigen-specific CD4+ T cell responses, including high Th1 and T follicular helper (Tfh) responses. Interestingly, in the absence of elastase activity, mucosal SIgA responses were induced after systemic immunization with alum as adjuvant. Importantly, lack or suppression of elastase activity enhanced the magnitude of anti–SARS-CoV-2 spike subunit 1 (S1) antibodies, and these antibodies reacted with the same epitopes of spike 1 protein as sera from COVID-19 patients. Therefore, suppression of neutrophil elastase could represent an attractive strategy for improving the efficacy of alum-based injected vaccines for the induction of broad immunity, including mucosal immunity.
Collapse
|
88
|
Lin YJ, Wei KC, Chen PY, Lim M, Hwang TL. Roles of Neutrophils in Glioma and Brain Metastases. Front Immunol 2021; 12:701383. [PMID: 34484197 PMCID: PMC8411705 DOI: 10.3389/fimmu.2021.701383] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/19/2021] [Indexed: 12/16/2022] Open
Abstract
Neutrophils, which are the most abundant circulating leukocytes in humans, are the first line of defense against bacterial and fungal infections. Recent studies have reported the role and importance of neutrophils in cancers. Glioma and brain metastases are the most common malignant tumors of the brain. The tumor microenvironment (TME) in the brain is complex and unique owing to the brain-blood barrier or brain-tumor barrier, which may prevent drug penetration and decrease the efficacy of immunotherapy. However, there are limited studies on the correlation between brain cancer and neutrophils. This review discusses the origin and functions of neutrophils. Additionally, the current knowledge on the correlation between neutrophil-to-lymphocyte ratio and prognosis of glioma and brain metastases has been summarized. Furthermore, the implications of tumor-associated neutrophil (TAN) phenotypes and the functions of TANs have been discussed. Finally, the potential effects of various treatments on TANs and the ability of neutrophils to function as a nanocarrier of drugs to the brain TME have been summarized. However, further studies are needed to elucidate the complex interactions between neutrophils, other immune cells, and brain tumor cells.
Collapse
Affiliation(s)
- Ya-Jui Lin
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Taiwan
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou, Taiwan
- Department of Neurosurgery, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, New Taipei, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Pin-Yuan Chen
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurosurgery, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Michael Lim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
- Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| |
Collapse
|
89
|
Recent trends of NFκB decoy oligodeoxynucleotide-based nanotherapeutics in lung diseases. J Control Release 2021; 337:629-644. [PMID: 34375688 DOI: 10.1016/j.jconrel.2021.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 02/07/2023]
Abstract
Nuclear factor κB (NFκB) is a unique protein complex that plays a major role in lung inflammation and respiratory dysfunction. The NFκB signaling pathway, therefore becomes an avenue for the development of potential pharmacological interventions, especially in situations where chronic inflammation is often constitutively active and plays a key role in the pathogenesis and progression of the disease. NFκB decoy oligodeoxynucleotides (ODNs) are double-stranded and carry NFκB binding sequences. They prevent the formation of NFκB-mediated inflammatory cytokines and thus have been employed in the treatment of a variety of chronic inflammatory diseases. However, the systemic administration of naked decoy ODNs restricts their therapeutic effectiveness because of their poor pharmacokinetic profile, instability, degradation by cellular enzymes and their low cellular uptake. Both structural modification and nanotechnology have shown promising results in enhancing the pharmacokinetic profiles of potent therapeutic substances and have also shown great potential in the treatment of respiratory diseases such as asthma, chronic obstructive pulmonary disease and cystic fibrosis. In this review, we examine the contribution of NFκB activation in respiratory diseases and recent advancements in the therapeutic use of decoy ODNs. In addition, we also highlight the limitations and challenges in use of decoy ODNs as therapeutic molecules, cellular uptake of decoy ODNs, and the current need for novel delivery systems to provide efficient delivery of decoy ODNs. Furthermore, this review provides a common platform for discussion on the existence of decoy ODNs, as well as outlining perspectives on the latest generation of delivery systems that encapsulate decoy ODNs and target NFκB in respiratory diseases.
Collapse
|
90
|
Sun X, Wang D, Wang Y, Ye L, Shi F, Qu S, Liang H, Zen K. PKM2 controls the degranulation of secondary and tertiary granules in neutrophils by phosphorylating SNAP-23. Cell Mol Immunol 2021; 18:2048-2050. [PMID: 34155337 PMCID: PMC8322427 DOI: 10.1038/s41423-021-00718-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/21/2022] Open
Affiliation(s)
- Xinlei Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Dong Wang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yan Wang
- School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Lei Ye
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fujie Shi
- School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Shuang Qu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
- School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Hongwei Liang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ke Zen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China.
- School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
91
|
Wei R, Li X, Wang X, Wang Y, Zhang X, Zhang N, Wang J, Yang J, Zhang X, Gong P, Li J. Trypanosoma evansi triggered neutrophil extracellular traps formation dependent on myeloperoxidase, neutrophil elastase, and extracellular signal-regulated kinase 1/2 signaling pathways. Vet Parasitol 2021; 296:109502. [PMID: 34214944 DOI: 10.1016/j.vetpar.2021.109502] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/06/2021] [Accepted: 06/16/2021] [Indexed: 10/21/2022]
Abstract
Trypanosoma evansi infects a wide range of hosts to cause huge economic losses in livestock industry. In recent years, it has been demonstrated that neutrophils extracellular traps (NETs) play a critical role in combating parasite infections. However, the role of NETs in the resistance to T. evansi infection is still unclear. In this study, T. evansi induced NETs were observed and their components were determined. The effect of NETs on the viability and motility of T. evansi were estimated. The production of reactive oxygen species (ROS) and Lactate dehydrogenase (LDH) activity in the process of T. evansi-induced NETs formation were detected. The effect of ERK1/2 signaling pathway, neutrophil elastase (NE), myeloperoxidase (MPO), store-operated Ca(2+) entry (SOCE) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase on T. evansi triggered NETs formation were determined. The results showed that neutrophils could release ETs after being stimulated with T. evansi and the structures of NETs mainly consisted of DNA decorated with histone 3 (H3), NE, and MPO. NETs could reduce the parasite motility without affecting the parasite viability. T. evansi-induced NETs formation was dose and time-dependent and was accompanied by ROS production. Inhibitor assays suggested that the formation of NETs induced by T. evansi was dependent on MPO, NE and ERK1/2 signaling pathway but independent on NADPH oxidase and SOCE. In addition, there was no significant changes in LDH activity during NETs formation. This study is the first report of T. evansi-induced NETs formation.
Collapse
Affiliation(s)
- Ran Wei
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Xin Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Xiaocen Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Yuru Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Xu Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Nan Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Jingsen Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Ju Yang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Xichen Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Pengtao Gong
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| | - Jianhua Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 5333 Xian Road, Changchun, 130062, Jilin, China.
| |
Collapse
|
92
|
Mok AC, Mody CH, Li SS. Immune Cell Degranulation in Fungal Host Defence. J Fungi (Basel) 2021; 7:484. [PMID: 34208679 PMCID: PMC8234259 DOI: 10.3390/jof7060484] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/08/2021] [Accepted: 06/11/2021] [Indexed: 12/13/2022] Open
Abstract
Humans have developed complex immune systems that defend against invading microbes, including fungal pathogens. Many highly specialized cells of the immune system share the ability to store antimicrobial compounds in membrane bound organelles that can be immediately deployed to eradicate or inhibit growth of invading pathogens. These membrane-bound organelles consist of secretory vesicles or granules, which move to the surface of the cell, where they fuse with the plasma membrane to release their contents in the process of degranulation. Lymphocytes, macrophages, neutrophils, mast cells, eosinophils, and basophils all degranulate in fungal host defence. While anti-microbial secretory vesicles are shared among different immune cell types, information about each cell type has emerged independently leading to an uncoordinated and confusing classification of granules and incomplete description of the mechanism by which they are deployed. While there are important differences, there are many similarities in granule morphology, granule content, stimulus for degranulation, granule trafficking, and release of granules against fungal pathogens. In this review, we describe the similarities and differences in an attempt to translate knowledge from one immune cell to another that may facilitate further studies in the context of fungal host defence.
Collapse
Affiliation(s)
- Adley Ch Mok
- Department of Microbiology Immunology and Infectious Diseases, Cumming School of Medicine, University Calgary, Calgary, AB T2N 4N1, Canada
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Christopher H Mody
- Department of Microbiology Immunology and Infectious Diseases, Cumming School of Medicine, University Calgary, Calgary, AB T2N 4N1, Canada
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Shu Shun Li
- Department of Microbiology Immunology and Infectious Diseases, Cumming School of Medicine, University Calgary, Calgary, AB T2N 4N1, Canada
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
93
|
Gu HJ, Li MF, Sun L. A deep-sea pathogenic Bacillus subtilis isolate employs different strategies to escape the killing of teleost and murine complements. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 119:104037. [PMID: 33545212 DOI: 10.1016/j.dci.2021.104037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 06/12/2023]
Abstract
Bacillus subtilis subsp. subtilis G7 was isolated from a deep-sea hydrothermal vent and is pathogenic to pathogenic to fish (Japanese flounder) and mice. G7 is able to survive in host sera and phagocytes. In this study, we investigated the underlying mechanism of G7 serum resistance. We found that (i) the remaining complement activity was very low in G7-incubated flounder serum but high in G7-incubated mouse serum; (ii) cleaved C3 and C5 components were detected on flounder serum-incubated G7 but not on mouse serum-incubated G7; (iii) abundant uncleaved C5 was localized in G7-incubated mouse, but not flounder, serum; (iv) G7-incubated flounder, but not mouse, serum exhibited strong chemotactic activity; (v) pre-treatment with low-dose lysozyme abolished the serum resistance of G7. Hence, G7 activates flounder complement but is protected from complement-mediated destruction by its cell wall structure, while G7 prevents the activation of mouse complement. These results indicate that G7 employs different mechanisms to avoid the complement killing of different hosts.
Collapse
Affiliation(s)
- Han-Jie Gu
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Institute of Tropical Bioscience and Biotechnology, Hainan Academy of Tropical Agricultural Resource, CATAS, Haikou, China
| | - Mo-Fei Li
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Li Sun
- CAS Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao, China; Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology, Qingdao, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
94
|
Charles Jacob HK, Charles Richard JL, Signorelli R, Kashuv T, Lavania S, Vaish U, Boopathy R, Middleton A, Boone MM, Sundaram R, Dudeja V, Saluja AK. Modulation of Early Neutrophil Granulation: The Circulating Tumor Cell-Extravesicular Connection in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13112727. [PMID: 34072942 PMCID: PMC8198339 DOI: 10.3390/cancers13112727] [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: 04/13/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Circulating tumor cells (CTCs) found in the blood of pancreatic cancer patients show a worse prognosis to therapy if they are seen in clusters of cells with neutrophils or platelets or with other cell types than when they are seen as singlets. We wanted to investigate if there is a secondary mode of communication between the CTCs and neutrophils that causes them to associate. We describe for the first time an extravesicular (EV) mediated communication between CTCs and neutrophils that modulates early transcriptome changes that can cause neutrophils to partially degranulate and form associations. We also identify the protein cargo carried in such EVs and how when added to naïve neutrophils, they can modulate transcriptomic changes in neutrophils partially disarming them to form clusters rather than undergo specialized cell death, which is characterized by release of condensed chromatin (NETs) and granular contents termed as NETosis. Abstract Tumor cells dissociate from the primary site and enter into systemic circulation (circulating tumor cells, CTCs) either alone or as tumor microemboli (clusters); the latter having an increased predisposition towards forming distal metastases than single CTCs. The formation of clusters is, in part, created by contacts between cell–cell junction proteins and/or cytokine receptor pairs with other cells such as neutrophils, platelets, fibroblasts, etc. In the present study, we provide evidence for an extravesicular (EV) mode of communication between pancreatic cancer CTCs and neutrophils. Our results suggest that the EV proteome of CTCs contain signaling proteins that can modulate degranulation and granule mobilization in neutrophils and, also, contain tissue plasminogen activator and other proteins that can regulate cluster formation. By exposing naïve neutrophils to EVs isolated from CTCs, we further show how these changes are modulated in a dynamic fashion indicating evidence for a deeper EV based remodulatory effect on companion cells in clusters.
Collapse
Affiliation(s)
- Harrys Kishore Charles Jacob
- Departments of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (H.K.C.J.); (S.L.); (A.M.); (R.S.); (V.D.)
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL 33136, USA
| | - John Lalith Charles Richard
- School of Biosciences, Engineering and Technology (SBET), VIT Bhopal University, Madhya Pradesh 466114, India;
| | | | - Tyler Kashuv
- Department of Biochemistry and Molecular Biology, University of Miami, Miami, FL 33146, USA;
| | - Shweta Lavania
- Departments of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (H.K.C.J.); (S.L.); (A.M.); (R.S.); (V.D.)
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Utpreksha Vaish
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35233, USA;
| | - Ranjitha Boopathy
- Department of Life Sciences, Shiv Nadar University, Greater Noida 201304, India;
| | - Ashley Middleton
- Departments of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (H.K.C.J.); (S.L.); (A.M.); (R.S.); (V.D.)
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL 33136, USA
| | | | - Ramakrishnan Sundaram
- Departments of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (H.K.C.J.); (S.L.); (A.M.); (R.S.); (V.D.)
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL 33136, USA
| | - Vikas Dudeja
- Departments of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (H.K.C.J.); (S.L.); (A.M.); (R.S.); (V.D.)
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL 33136, USA
- Department of Life Sciences, Shiv Nadar University, Greater Noida 201304, India;
| | - Ashok Kumar Saluja
- Departments of Surgery, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; (H.K.C.J.); (S.L.); (A.M.); (R.S.); (V.D.)
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL 33136, USA
- Correspondence: ; Tel.: +1-305-243-2703
| |
Collapse
|
95
|
Han Y, Liu Y, Ma X, Shen A, Liu Y, Weeranoppanant N, Dong H, Li Y, Ren T, Kuai L, Li B, An M, Li Y. Antibiotics armed neutrophils as a potential therapy for brain fungal infection caused by chemotherapy-induced neutropenia. Biomaterials 2021; 274:120849. [PMID: 34022739 DOI: 10.1016/j.biomaterials.2021.120849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 04/09/2021] [Accepted: 04/19/2021] [Indexed: 12/17/2022]
Abstract
Chemotherapy-induced neutropenia, a symptom of neutrophil depletion, makes cancer patients highly susceptible to invasive fungal infection with substantial morbidity and mortality. To address the cryptococcal brain infection in this condition, this study attempts to arm neutrophils (NEs) with antibiotics to potentiate the antifungal capability of NEs. To allow effective integration, amphotericin B, a potent antibiotic, is assembled with albumin nanoparticles through hydrophobic and hydrogen-bond interactions to form AmB@BSA nanoparticles (A-NPs). The nutrient composition (albumin) and virus-like size (~40 nm) facilitate efficient uptake of A-NPs by NEs to construct the antibiotics-armed NEs. It is demonstrated that the armed NEs can maintain the intrinsic biological functions of NEs, such as cell viability and capacity of migration to an inflammatory site. In a neutropenic mouse model of brain fungal infection, the treatment with the armed NEs allows for preventing fungal invasion more effectively than that with the native NEs, without the apparent systemic toxicity. Such a synergistic anti-infection system maximizes the antifungal effects by taking advantage of NEs and antibiotics. It provides a potential NEs-mediated therapeutic approach for treating fungal infection caused by chemotherapy-induced neutropenia.
Collapse
Affiliation(s)
- Yi Han
- Shanghai Tenth People's Hospital, Department of Medical Ultrasound, Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, PR China
| | - Yanchao Liu
- Department of Clinical Pharmacy, Shanghai General Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200080, PR China
| | - Xiaoyi Ma
- Shanghai Tenth People's Hospital, Department of Medical Ultrasound, Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, PR China
| | - Aijun Shen
- Department of Medical Imaging, Tongji Hospital, Tongji University, Shanghai, 200065, PR China
| | - Yiqiong Liu
- Shanghai Tenth People's Hospital, Department of Medical Ultrasound, Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, PR China
| | - Nopphon Weeranoppanant
- Department of Chemical Engineering, Burapha University, 169 Longhard Bangsaen, Saensook, Chonburi, 20131, Thailand
| | - Haiqing Dong
- Shanghai Tenth People's Hospital, Department of Medical Ultrasound, Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, PR China
| | - Yan Li
- Shanghai Tenth People's Hospital, Department of Medical Ultrasound, Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, PR China
| | - Tianbin Ren
- Shanghai Tenth People's Hospital, Department of Medical Ultrasound, Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, PR China
| | - Le Kuai
- Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai, 200092, PR China
| | - Bin Li
- Shanghai Dermatology Hospital, Tongji University School of Medicine, Shanghai, 200092, PR China.
| | - Maomao An
- Shanghai Tenth People's Hospital, Department of Medical Ultrasound, Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, PR China.
| | - Yongyong Li
- Shanghai Tenth People's Hospital, Department of Medical Ultrasound, Department of Pharmacology, Tongji University School of Medicine, Shanghai, 200092, PR China.
| |
Collapse
|
96
|
Misiukiewicz-Stepien P, Paplinska-Goryca M. Biological effect of PM 10 on airway epithelium-focus on obstructive lung diseases. Clin Immunol 2021; 227:108754. [PMID: 33964432 DOI: 10.1016/j.clim.2021.108754] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 04/16/2021] [Accepted: 05/03/2021] [Indexed: 12/11/2022]
Abstract
Recently, a continuous increase in environmental pollution has been observed. Despite wide-scale efforts to reduce air pollutant emissions, the problem is still relevant. Exposure to elevated levels of airborne particles increased the incidence of respiratory diseases. PM10 constitute the largest fraction of air pollutants, containing particles with a diameter of less than 10 μm, metals, pollens, mineral dust and remnant material from anthropogenic activity. The natural airway defensive mechanisms against inhaled material, such as mucus layer, ciliary clearance and macrophage phagocytic activity, may be insufficient for proper respiratory function. The epithelium layer can be disrupted by ongoing oxidative stress and inflammatory processes induced by exposure to large amounts of inhaled particles as well as promote the development and exacerbation of obstructive lung diseases. This review draws attention to the current state of knowledge about the physical features of PM10 and its impact on airway epithelial cells, and obstructive pulmonary diseases.
Collapse
Affiliation(s)
- Paulina Misiukiewicz-Stepien
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland; Department of Internal Medicine, Pulmonary Diseases and Allergy, Medical University of Warsaw, Poland.
| | | |
Collapse
|
97
|
MicroRNA: Could It Play a Role in Bovine Endometritis? Inflammation 2021; 44:1683-1695. [PMID: 33907916 DOI: 10.1007/s10753-021-01458-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/15/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Endometritis in dairy cows is a major economic problem worldwide; without advances in lifestyle management and drug treatment, it causes high morbidity and death. Micro ribonucleic acid (miRNAs) these days is seen as an important part of gene control networks. It is a class of small nucleotides 20-25, single-stranded RNA molecules. In endometritis, the inflammatory response caused by the gram-negative bacteria Escherichia coli (E. coli) alters the expression of miRNA which can regulate the innate immune system. This manuscript reviews (1) the interaction of miRNAs with the signaling of NF-κB and dysregulation of miRNAs and NF-κB activity in endometritis and (2) the activity of miR-let-7c, miR-148a, and miR-488 in NF-κB activation and their effect on endometritis. Cows with reduced immunity are more vulnerable to transition diseases, such as endometritis. During post-partum, cows undergo stress, metabolic disorders, hormonal imbalance, negative energy balance, and changes in diet. One of the many categories of regulatory molecules, which explain its natural function and pathological impact on NF-κB dysregulation, is important to inform the complexity of the immune system and to develop treatments for endometritis. It shows that miRNAs could have multiple applications in veterinary medicine. Nevertheless, a comprehensive study of is essential which should be aimed at exploring the role of microRNA at physiological level and its effect due to dysfunction and dysregulation.
Collapse
|
98
|
Zhao Y, Cai C, Samir J, Palgen JL, Keoshkerian E, Li H, Bull RA, Luciani F, An H, Lloyd AR. Human CD8 T-stem cell memory subsets phenotypic and functional characterization are defined by expression of CD122 or CXCR3. Eur J Immunol 2021; 51:1732-1747. [PMID: 33844287 DOI: 10.1002/eji.202049057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/07/2021] [Accepted: 04/07/2021] [Indexed: 11/12/2022]
Abstract
Long-lived T-memory stem cells (TSCM ) are key to both naturally occurring and vaccine-conferred protection against infection. These cells are characterized by the CD45RA+ CCR7+ CD95+ phenotype. Significant heterogeneity within the TSCM population is recognized, but distinguishing surface markers and functional characterization of potential subsets are lacking. Human CD8 TSCM subsets were identified in healthy subjects who had been previously exposed to CMV or Influenza (Flu) virus in flow cytometry by expression of CD122 or CXCR3, and then characterized in proliferation, multipotency, self-renewal, and intracellular cytokine production (TNF-α, IL-2, IFN-γ), together with transcriptomic profiles. The TSCM CD122hi -expressing subset (versus CD122lo ) demonstrated greater proliferation, greater multipotency, and enhanced polyfunctionality with higher frequencies of triple positive (TNF-α, IL-2, IFN-γ) cytokine-producing cells upon exposure to recall antigen. The TSCM CXCR3lo subpopulation also had increased proliferation and polyfunctional cytokine production. Transcriptomic analysis further showed that the TSCM CD122hi population had increased expression of activation and homing molecules, such as Ccr6, Cxcr6, Il12rb, and Il18rap, and downregulated cell proliferation inhibitors, S100A8 and S100A9. These data reveal that the TSCM CD122hi phenotype is associated with increased proliferation, enhanced multipotency and polyfunctionality with an activated memory-cell like transcriptional profile, and hence, may be favored for induction by immunization and for adoptive immunotherapy.
Collapse
Affiliation(s)
- Yanran Zhao
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Curtis Cai
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jerome Samir
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jean-Louis Palgen
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Elizabeth Keoshkerian
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Hui Li
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Rowena A Bull
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Fabio Luciani
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia.,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Hongyan An
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Andrew R Lloyd
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
99
|
Blanch-Ruiz MA, Ortega-Luna R, Martínez-Cuesta MÁ, Álvarez Á. The Neutrophil Secretome as a Crucial Link between Inflammation and Thrombosis. Int J Mol Sci 2021; 22:4170. [PMID: 33920656 PMCID: PMC8073391 DOI: 10.3390/ijms22084170] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular diseases are a leading cause of death. Blood-cell interactions and endothelial dysfunction are fundamental in thrombus formation, and so further knowledge of the pathways involved in such cellular crosstalk could lead to new therapeutical approaches. Neutrophils are secretory cells that release well-known soluble inflammatory signaling mediators and other complex cellular structures whose role is not fully understood. Studies have reported that neutrophil extracellular vesicles (EVs) and neutrophil extracellular traps (NETs) contribute to thrombosis. The objective of this review is to study the role of EVs and NETs as key factors in the transition from inflammation to thrombosis. The neutrophil secretome can promote thrombosis due to the presence of different factors in the EVs bilayer that can trigger blood clotting, and to the release of soluble mediators that induce platelet activation or aggregation. On the other hand, one of the main pathways by which NETs induce thrombosis is through the creation of a scaffold to which platelets and other blood cells adhere. In this context, platelet activation has been associated with the induction of NETs release. Hence, the structure and composition of EVs and NETs, as well as the feedback mechanism between the two processes that causes pathological thrombus formation, require exhaustive analysis to clarify their role in thrombosis.
Collapse
Affiliation(s)
- María Amparo Blanch-Ruiz
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - Raquel Ortega-Luna
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
| | - María Ángeles Martínez-Cuesta
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (M.A.B.-R.); (R.O.-L.)
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBERehd), 46010 Valencia, Spain
| |
Collapse
|
100
|
Liu G, Yuan C, Ma J, Pan Y, Xu H. Influence of Immune Microenvironment on Diagnosis and Prognosis of Head and Neck Squamous Cell Carcinoma. Front Oncol 2021; 11:604784. [PMID: 33816236 PMCID: PMC8010259 DOI: 10.3389/fonc.2021.604784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/27/2021] [Indexed: 12/26/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an immunosuppressive malignancy accompanied by noted alterations in various immune cells and cytokines. Recognition of the immune system's role in contributing to cancer development is an important advancement in our original understanding of carcinoma. We obtained HNSCC gene expression and clinical data from The Cancer Genome Atlas (TCGA) database. We assessed the relative proportion of 22 Infiltrating immune cell types in both HNSCC and adjacent non-cancer tissues using Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) method, identifying the influence of the immune cells content in tumor staging and survival prediction. We further predicted the tumor purity, and the presence of infiltrating stromal/immune cells in HNSCC tissues using Estimation of STromal and Immune cells in Malignant Tumor tissues using Expression data (ESTIMATE) algorithm, identifying its potential correlation with patient survival. Stromal and immune score-associated differentially expressed genes (DEGs) were subsequently verified and their roles in immune response were displayed by functional enrichment analysis and protein-protein interaction (PPI) network. Our research demonstrated the underlying association between the immune microenvironment and HNSCC, and the results were intended to serve as valuable terms for HNSCC diagnosis, prognosis, and targeted immune therapy.
Collapse
Affiliation(s)
- Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Chunjue Yuan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jiaojiao Ma
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Haibo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|