1
|
Xue H, Xie R, Wang Z, Fan W, Wei Y, Zhang L, Zhao D, Song Z. Coordination of Neutrophil and Apoptosis-Inducing Ligand in Inflammatory Diseases. J Inflamm Res 2025; 18:3607-3621. [PMID: 40099000 PMCID: PMC11911651 DOI: 10.2147/jir.s506807] [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: 11/15/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
As the most abundant innate immune cells, neutrophils play a key role in host's anti-infective activity and tissue damage/repair process of sterile inflammation. Due to the restriction of apoptosis and other regulatory mechanisms, neutrophils have a short survival time in vivo. Because of the death domain of cytoplasmic regions, some members of tumor necrosis factor receptor superfamily (TNFRSF) are defined as death receptors, such as TNFR-I, Fas and DR4/DR5. TNF-α, FasL and TRAIL, which are known as apoptosis-inducing ligand, can bind to death receptors and activate intracellular apoptosis pathways to induce apoptosis. Accumulating studies found that these three apoptosis-inducing ligands play an important role in the immune system by coordinating with neutrophil, which including neutrophil recruitment/infiltration and function performing. In this review, we summarize existing studies targeting neutrophils as diagnosis and treatment for diseases, and focus on the involvement of neutrophils which regulated by apoptosis-inducing ligands in inflammatory diseases under current cognition.
Collapse
Affiliation(s)
- Hanyu Xue
- The First Affiliated Hospital of Henan University, School of Medicine, Henan University, Kaifeng, 475004, People's Republic of China
| | - Ran Xie
- School of Medical Technology, Shangqiu Medical College, Shangqiu, 476000, People's Republic of China
| | - Zhiwei Wang
- Department of Oncology, The First Affiliated Hospital of Henan University, Kaifeng, 475000, People's Republic of China
| | - Wenqian Fan
- School of Medicine, Henan University, Kaifeng, 475004, People's Republic of China
| | - Yinxiang Wei
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Lijie Zhang
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Dan Zhao
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Zhiming Song
- Department of Cardiology, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
- Kaifeng Key Laboratory for Modulation and Rehabilitation of Cardiac Function, The First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| |
Collapse
|
2
|
Zhao H, Lin Z, Zhang P, Rao J, Xu S, Luo Q, Li J. KLF2 controls the apoptosis of neutrophils and is associated with disease activity of systemic lupus erythematosus. Arthritis Res Ther 2024; 26:222. [PMID: 39702240 DOI: 10.1186/s13075-024-03461-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Neutropenia is more common in patients with systemic lupus erythematosus (SLE) and is a major cause of life-threatening infections. The increased apoptosis of neutrophils is likely to be an essential cause of neutropenia in SLE. However, the detailed mechanisms of increased neutrophil apoptosis in SLE remain unknown. This study focused on the role of Krüppel-like factor 2 (KLF2) in the regulation of neutrophil apoptosis and its association with SLE disease activity. METHODS The levels of KLF2 in neutrophils from SLE patients and healthy controls (HCs) were detected by RT-PCR and western blot. The relationship between the levels of KLF2 and the apoptosis levels of neutrophils in SLE patients was analyzed. The KLF2 inhibitor Geranylgeranyl pyrophosphate (GGPP) and the KLF2 inducer geranylgeranyl transferase inhibitor (GGTI-298) were used to incubate with neutrophils to investigate the role of KLF2 in the regulation of neutrophil apoptosis. To clarify whether serum from SLE patients affects neutrophil KLF2 expression and apoptosis, sera from SLE patients were collected and used to incubate with neutrophils from HCs, followed by the detection of KLF2 levels and apoptosis levels of neutrophils. Additionally, the correlation between KLF2 levels and SLE disease activity index (SLEDAI) was analyzed. RESULTS The expression of KLF2 in neutrophils of SLE patients was significantly suppressed, and the decreased KLF2 was associated with the upregulation of neutrophil apoptosis. Moreover, newly diagnosed SLE patients, SLE patients with higher serum IgG and positive anti-Smith antibodies had lower KLF2 expression. Furthermore, we demonstrated that modulating the expression of KLF2 can regulate the apoptosis of neutrophils. The levels of KLF2 in neutrophils were associated with the SLEDAI. In addition, we found that serum from SLE patients could induce apoptosis in neutrophils by down-regulating the expression of KLF2. CONCLUSIONS KLF2 controls the apoptosis of neutrophils and is associated with SLEDAI, which suggests that KLF2 in neutrophils may be involved in the occurrence and development of SLE.
Collapse
Affiliation(s)
- Hongshuai Zhao
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Zaichuan Lin
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Peiwen Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Jiayue Rao
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Shumin Xu
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Qing Luo
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Nanchang Key Laboratory of Diagnosis of Infectious Diseases, Nanchang, Jiangxi, 330006, China.
- Institute of infection and immunity, Nanchang University, Nanchang, 330006, Jiangxi, China.
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No.17, YongwaiZhengjie, Donghu District, Nanchang, Jiangxi, P.R. China.
| | - Junming Li
- Department of Clinical Laboratory, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
- Institute of infection and immunity, Nanchang University, Nanchang, 330006, Jiangxi, China.
- Medical Center of Burn plastic and wound repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi, 330006, China.
- Department of Clinical Laboratory, The First Affiliated Hospital of Nanchang University, No.17, YongwaiZhengjie, Donghu District, Nanchang, Jiangxi, P.R. China.
| |
Collapse
|
3
|
Yipeng Z, Chao C, Ranran L, Tingting P, Hongping Q. Metabolism: a potential regulator of neutrophil fate. Front Immunol 2024; 15:1500676. [PMID: 39697327 PMCID: PMC11652355 DOI: 10.3389/fimmu.2024.1500676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Neutrophils are essential components of the innate immune system that defend against the invading pathogens, such as bacteria, viruses, and fungi, as well as having regulatory roles in various conditions, including tissue repair, cancer immunity, and inflammation modulation. The function of neutrophils is strongly related to their mode of cell death, as different types of cell death involve various cellular and molecular alterations. Apoptosis, a non-inflammatory and programmed type of cell death, is the most common in neutrophils, while other modes of cell death, including NETOsis, necrosis, necroptosis, autophagy, pyroptosis, and ferroptosis, have specific roles in neutrophil function regulation. Immunometabolism refers to energy and substance metabolism in immune cells, and profoundly influences immune cell fate and immune system function. Intercellular and intracellular signal transduction modulate neutrophil metabolism, which can, in turn, alter their activities by influencing various cell signaling pathways. In this review, we compile an extensive body of evidence demonstrating the role of neutrophil metabolism in their various forms of cell death. The review highlights the intricate metabolic characteristics of neutrophils and their interplay with various types of cell death.
Collapse
Affiliation(s)
| | | | | | - Pan Tingting
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China
| | - Qu Hongping
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Grayson KA, Greenlee JD, Himmel LE, Hapach LA, Reinhart-King CA, King MR. Spatial distribution of tumor-associated macrophages in an orthotopic prostate cancer mouse model. Pathol Oncol Res 2024; 30:1611586. [PMID: 38689823 PMCID: PMC11058651 DOI: 10.3389/pore.2024.1611586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/03/2024] [Indexed: 05/02/2024]
Abstract
Mounting evidence suggests that the immune landscape within prostate tumors influences progression, metastasis, treatment response, and patient outcomes. In this study, we investigated the spatial density of innate immune cell populations within NOD.SCID orthotopic prostate cancer xenografts following microinjection of human DU145 prostate cancer cells. Our laboratory has previously developed nanoscale liposomes that attach to leukocytes via conjugated E-selectin (ES) and kill cancer cells via TNF-related apoptosis inducing ligand (TRAIL). Immunohistochemistry (IHC) staining was performed on tumor samples to identify and quantify leukocyte infiltration for different periods of tumor growth and E-selectin/TRAIL (EST) liposome treatments. We examined the spatial-temporal dynamics of three different immune cell types infiltrating tumors using QuPath image analysis software. IHC staining revealed that F4/80+ tumor-associated macrophages (TAMs) were the most abundant immune cells in all groups, irrespective of time or treatment. The density of TAMs decreased over the course of tumor growth and decreased in response to EST liposome treatments. Intratumoral versus marginal analysis showed a greater presence of TAMs in the marginal regions at 3 weeks of tumor growth which became more evenly distributed over time and in tumors treated with EST liposomes. TUNEL staining indicated that EST liposomes significantly increased cell apoptosis in treated tumors. Additionally, confocal microscopy identified liposome-coated TAMs in both the core and periphery of tumors, highlighting the ability of liposomes to infiltrate tumors by "piggybacking" on macrophages. The results of this study indicate that TAMs represent the majority of innate immune cells within NOD.SCID orthotopic prostate tumors, and spatial density varies widely as a function of tumor size, duration of tumor growth, and treatment of EST liposomes.
Collapse
Affiliation(s)
- Korie A. Grayson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Joshua D. Greenlee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Lauren E. Himmel
- Department of Pathology, Microbiology and Immunology, Translational Pathology Shared Resource, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Lauren A. Hapach
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | | | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
5
|
Bodac A, Mayet A, Rana S, Pascual J, Bowler AD, Roh V, Fournier N, Craciun L, Demetter P, Radtke F, Meylan E. Bcl-xL targeting eliminates ageing tumor-promoting neutrophils and inhibits lung tumor growth. EMBO Mol Med 2024; 16:158-184. [PMID: 38177532 PMCID: PMC10897164 DOI: 10.1038/s44321-023-00013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 01/06/2024] Open
Abstract
Elevated peripheral blood and tumor-infiltrating neutrophils are often associated with a poor patient prognosis. However, therapeutic strategies to target these cells are difficult to implement due to the life-threatening risk of neutropenia. In a genetically engineered mouse model of lung adenocarcinoma, tumor-associated neutrophils (TAN) demonstrate tumor-supportive capacities and have a prolonged lifespan compared to circulating neutrophils. Here, we show that tumor cell-derived GM-CSF triggers the expression of the anti-apoptotic Bcl-xL protein and enhances neutrophil survival through JAK/STAT signaling. Targeting Bcl-xL activity with a specific BH3 mimetic, A-1331852, blocked the induced neutrophil survival without impacting their normal lifespan. Specifically, oral administration with A-1331852 decreased TAN survival and abundance, and reduced tumor growth without causing neutropenia. We also show that G-CSF, a drug used to combat neutropenia in patients receiving chemotherapy, increased the proportion of young TANs and augmented the anti-tumor effect resulting from Bcl-xL blockade. Finally, our human tumor data indicate the same role for Bcl-xL on pro-tumoral neutrophil survival. These results altogether provide preclinical evidence for safe neutrophil targeting based on their aberrant intra-tumor longevity.
Collapse
Affiliation(s)
- Anita Bodac
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Abdullah Mayet
- Laboratory of Immunobiology, Department of Molecular Biology, Université libre de Bruxelles, 6041, Gosselies, Belgium
- Lung Cancer and Immuno-Oncology laboratory, Bordet Cancer Research Laboratories, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université libre de Bruxelles, 1070, Bruxelles, Belgium
- ULB Cancer Research Center (U-CRC) and ULB Center for Research in Immunology (U-CRI), 1070, Bruxelles, Belgium
| | - Sarika Rana
- Laboratory of Immunobiology, Department of Molecular Biology, Université libre de Bruxelles, 6041, Gosselies, Belgium
- Lung Cancer and Immuno-Oncology laboratory, Bordet Cancer Research Laboratories, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université libre de Bruxelles, 1070, Bruxelles, Belgium
- ULB Cancer Research Center (U-CRC) and ULB Center for Research in Immunology (U-CRI), 1070, Bruxelles, Belgium
| | - Justine Pascual
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, 1015, Lausanne, Switzerland
| | - Amber D Bowler
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Vincent Roh
- Translational Data Science - Facility, SIB Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
- Agora Cancer Research Center, 1005, Lausanne, Switzerland
| | - Nadine Fournier
- Translational Data Science - Facility, SIB Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
- Agora Cancer Research Center, 1005, Lausanne, Switzerland
| | - Ligia Craciun
- Department of Pathology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université libre de Bruxelles, 1070, Bruxelles, Belgium
| | - Pieter Demetter
- Department of Pathology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université libre de Bruxelles, 1070, Bruxelles, Belgium
| | - Freddy Radtke
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Etienne Meylan
- Laboratory of Immunobiology, Department of Molecular Biology, Université libre de Bruxelles, 6041, Gosselies, Belgium.
- Lung Cancer and Immuno-Oncology laboratory, Bordet Cancer Research Laboratories, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université libre de Bruxelles, 1070, Bruxelles, Belgium.
- ULB Cancer Research Center (U-CRC) and ULB Center for Research in Immunology (U-CRI), 1070, Bruxelles, Belgium.
| |
Collapse
|
6
|
Li Y, Wu Y, Huang J, Cao X, An Q, Peng Y, Zhao Y, Luo Y. A variety of death modes of neutrophils and their role in the etiology of autoimmune diseases. Immunol Rev 2024; 321:280-299. [PMID: 37850797 DOI: 10.1111/imr.13284] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Neutrophils are important in the context of innate immunity and actively contribute to the progression of diverse autoimmune disorders. Distinct death mechanisms of neutrophils may exhibit specific and pivotal roles in autoimmune diseases and disease pathogenesis through the orchestration of immune homeostasis, the facilitation of autoantibody production, the induction of tissue and organ damage, and the incitement of pathological alterations. In recent years, more studies have provided in-depth examination of various neutrophil death modes, revealing nuances that challenge conventional understanding and underscoring their potential clinical utility in diagnosis and treatment. This review explores the multifaceted processes and characteristics of neutrophil death, with a focus on tailored investigations within various autoimmune diseases. It also highlights the potential interplay between neutrophil death and the landscape of autoimmune disorders. The review encapsulates the pertinent pathways implicated in various neutrophil death mechanisms across diverse autoimmune diseases while also charts possible avenues for future research.
Collapse
Affiliation(s)
- Yanhong Li
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yinlan Wu
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingang Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xue Cao
- Department of Rheumatology and Immunology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Qiyuan An
- School of Inspection and Biotechnology, Southern Medical University, Guangzhou, China
| | - Yun Peng
- Department of Rheumatology and Clinical Immunology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - Yi Zhao
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yubin Luo
- Department of Rheumatology & Immunology, Laboratory of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Xiao Y, Cheng Y, Liu WJ, Liu K, Wang Y, Xu F, Wang DM, Yang Y. Effects of neutrophil fate on inflammation. Inflamm Res 2023; 72:2237-2248. [PMID: 37925664 DOI: 10.1007/s00011-023-01811-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/18/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023] Open
Abstract
INTRODUCTION Neutrophils are important participants in the innate immune response. They rapidly and efficiently identify and clear infectious agents by expressing large numbers of membrane receptors. Upon tissue injury or pathogen invasion, neutrophils are the first immune cells to reach the site of injury and participate in the inflammatory response. MATERIALS AND METHODS A thorough search on PubMed related to neutrophil death or clearance pathways was performed. CONCLUSION Inflammatory response and tissue damage can be aggravated when neutrophils are not removed rapidly from the site of injury. Recent studies have shown that neutrophils can be cleared through a variety of pathways, including non-inflammatory and inflammatory death, as well as reverse migration. Non-inflammatory death pathways include apoptosis and autophagy. Inflammatory death pathways include necroptosis, pyroptosis and NETosis. This review highlights the basic properties of neutrophils and the impact of their clearance pathways on the inflammatory response.
Collapse
Affiliation(s)
- Yuan Xiao
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yang Cheng
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Wen-Jie Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Kun Liu
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yan Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Feng Xu
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - De-Ming Wang
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Yi Yang
- Department of Anesthesiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| |
Collapse
|
8
|
Wang JF, Sun BW, Marshall JC. Editorial: Neutrophil death regulation in critical illness. Front Immunol 2023; 14:1181056. [PMID: 37006251 PMCID: PMC10050726 DOI: 10.3389/fimmu.2023.1181056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Affiliation(s)
- Jia-feng Wang
- Faculty of Anesthesiology, Changhai Hospital, The Naval Medical University, Shanghai, China
| | - Bing-wei Sun
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Bing-wei Sun, ; John C. Marshall,
| | - John C. Marshall
- Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, Toronto, ON, Canada
- Department of Surgery, St Michael’s Hospital, Toronto, ON, Canada
- Department of Critical Care Medicine, St Michael’s Hospital, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- *Correspondence: Bing-wei Sun, ; John C. Marshall,
| |
Collapse
|
9
|
Koo S, Offner R, Haile SM, Brosig A, Hähnel V, Gruber M, Burkhardt R, Ahrens N. Granulocyte concentrate splitting does not affect phenotype and function. Transfusion 2023; 63:393-401. [PMID: 36519400 DOI: 10.1111/trf.17217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND More granulocyte concentrates (GCs) could be produced for more patients from the same donor if apheresis bags were split and stored for longer periods of time. Hence, we tested the hypothesis that splitting and extension of storage of GCs do not impair granulocyte function or viability. STUDY DESIGN AND METHODS Granulocyte apheresis concentrates were produced using modified fluid gelatin as a separation enhancer, split into two portions, and stored for 24 and 48 h. Granulocyte function, represented by cell migration, reactive oxygen species (ROS) production, and neutrophil extracellular trap formation (NETosis), was measured by live-cell imaging. ROS production, adhesive surface protein expression, and viability were measured by flow cytometry. RESULTS Splitting had no effect on any of the tested parameters. After 24 h of storage, live-cell imaging showed no significant difference in migration, time to maximum ROS production, time to half-maximum NETosis, viability, or CD11b expression, but ROS production induced by phorbol 12-myristate 13-acetate (PMA) decreased from an initial median fluorescence intensity of 1775-590 artificial units. After 48 h, PMA-induced ROS production, viability, and migration declined, as reflected by decreases in median total distance (119 vs. 63.5 μm) and median Euclidean distance (30.75 vs. 14.3 μm). CONCLUSION Splitting GC products has no effect on granulocyte viability or function, but extended storage >24 h does compromise granulocyte function. The findings confirm that GCs should be transfused within 24 h of collection. Longer storage cannot be recommended.
Collapse
Affiliation(s)
- Sebastian Koo
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Raubling, Germany.,Department of Anesthesiology, University Hospital Regensburg, Raubling, Germany
| | - Robert Offner
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Raubling, Germany
| | - Sophie-Marie Haile
- Department of Anesthesiology, University Hospital Regensburg, Raubling, Germany
| | - Andreas Brosig
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Raubling, Germany
| | - Viola Hähnel
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Raubling, Germany
| | - Michael Gruber
- Department of Anesthesiology, University Hospital Regensburg, Raubling, Germany
| | - Ralph Burkhardt
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Raubling, Germany
| | - Norbert Ahrens
- Institute for Clinical Chemistry and Laboratory Medicine, Transfusion Medicine, University Hospital Regensburg, Raubling, Germany.,Amedes MVZ for Laboratory Diagnostics, University Hospital Regensburg, Raubling, Germany
| |
Collapse
|
10
|
Mizuno Y, Shibata S, Ito Y, Taira H, Sugimoto E, Awaji K, Sato S. Interleukin-26–DNA complexes promote inflammation and dermal-epidermal separation in a modified human cryosection model of bullous pemphigoid. Front Immunol 2022; 13:1013382. [PMID: 36311716 PMCID: PMC9599390 DOI: 10.3389/fimmu.2022.1013382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Bullous pemphigoid (BP) is an autoimmune disease characterized by autoantibody-mediated activation of immune cells and subepidermal blister formation. Excess amounts of extracellular DNA are produced in BP, however, it remains unclear how extracellular DNA contributes to BP pathogenesis. Here we show a possible mechanism by which interleukin (IL)-26 binds to extracellular DNA released from neutrophils and eosinophils to support DNA sensing. Patients with BP exhibited high circulating levels of IL-26, forming IL-26–DNA complexes in the upper dermis and inside the blisters. IL-26–DNA complexes played a dual role in regulating local immunity and blister formation. First, they enhanced the production of inflammatory cytokines in monocytes and neutrophils. Second, and importantly, the complexes augmented the production and activity of proteases from co-cultured monocytes and neutrophils, which induced BP180 cleavage in keratinocytes and dermal-epidermal separation in a modified human cryosection model. Collectively, we propose a model in which IL-26 and extracellular DNA synergistically act on immune cells to enhance autoantibody-driven local immune responses and protease-mediated fragility of dermal-epidermal junction in BP.
Collapse
|
11
|
Landoni VI, Pittaluga JR, Carestia A, Castillo LA, Nebel MDC, Martire-Greco D, Birnberg-Weiss F, Schattner M, Schierloh P, Fernández GC. Neutrophil Extracellular Traps Induced by Shiga Toxin and Lipopolysaccharide-Treated Platelets Exacerbate Endothelial Cell Damage. Front Cell Infect Microbiol 2022; 12:897019. [PMID: 35811684 PMCID: PMC9262415 DOI: 10.3389/fcimb.2022.897019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/18/2022] [Indexed: 11/13/2022] Open
Abstract
Hemolytic uremic syndrome (HUS) is the most common cause of acute renal failure in the pediatric population. The etiology of HUS is linked to Gram-negative, Shiga toxin (Stx)-producing enterohemorrhagic bacterial infections. While the effect of Stx is focused on endothelial damage of renal glomerulus, cytokines induced by Stx or bacterial lipopolysaccharide (LPS) and polymorphonuclear cells (PMNs) are involved in the development of the disease. PMN release neutrophil extracellular traps (NETs) to eliminate pathogens, although NETs favor platelets (Plts) adhesion/thrombus formation and can cause tissue damage within blood vessels. Since thrombus formation and occlusion of vessels are characteristic of HUS, PMN–Plts interaction in the context of Stx may promote netosis and contribute to the endothelial damage observed in HUS. The aim of this study was to determine the relevance of netosis induced by Stx in the context of LPS-sensitized Plts on endothelial damage. We observed that Stx2 induced a marked enhancement of netosis promoted by Plts after LPS stimulation. Several factors seemed to promote this phenomenon. Stx2 itself increased the expression of its receptor on Plts, increasing toxin binding. Stx2 also increased LPS binding to Plts. Moreover, Stx2 amplified LPS induced P-selectin expression on Plts and mixed PMN–Plts aggregates formation, which led to activation of PMN enhancing dramatically NETs formation. Finally, experiments revealed that endothelial cell damage mediated by PMN in the context of Plts treated with LPS and Stx2 was decreased when NETs were disrupted or when mixed aggregate formation was impeded using an anti-P-selectin antibody. Using a murine model of HUS, systemic endothelial damage/dysfunction was decreased when NETs were disrupted, or when Plts were depleted, indicating that the promotion of netosis by Plts in the context of LPS and Stx2 plays a fundamental role in endothelial toxicity. These results provide insights for the first time into the pivotal role of Plts as enhancers of endothelial damage through NETs promotion in the context of Stx and LPS. Consequently, therapies designed to reduce either the formation of PMN–Plts aggregates or NETs formation could lessen the consequences of endothelial damage in HUS.
Collapse
Affiliation(s)
- Verónica Inés Landoni
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Jose R. Pittaluga
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Agostina Carestia
- Laboratorio de Trombosis Experimental e Inmunobiología de la Inflamación, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Luis Alejandro Castillo
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Marcelo de Campos Nebel
- Laboratorio de Mutagénesis, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Daiana Martire-Greco
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Federico Birnberg-Weiss
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Mirta Schattner
- Laboratorio de Trombosis Experimental e Inmunobiología de la Inflamación, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
| | - Pablo Schierloh
- Instituto de Investigación y Desarrollo en Bioingeniería y Bioinformática, Centro Científico Tecnológico Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Santa Fe, Argentina
| | - Gabriela C. Fernández
- Laboratorio de Fisiología de los Procesos Inflamatorios, Instituto de Medicina Experimental (IMEX)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)/Academia Nacional de Medicina de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Argentina
- *Correspondence: Gabriela C. Fernández, ;
| |
Collapse
|
12
|
Wang J, Lu S, Zheng K, He Z, Li W, Liu J, Guo N, Xie Y, Chen D, Xu M, Wu Y. Treponema pallidum delays the apoptosis of human polymorphonuclear neutrophils through the intrinsic and extrinsic pathways. Mol Immunol 2022; 147:157-169. [PMID: 35597181 DOI: 10.1016/j.molimm.2022.04.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/26/2022] [Accepted: 04/21/2022] [Indexed: 11/26/2022]
Abstract
Treponema pallidum is a "stealth pathogen" responsible for infectious sexually transmitted diseases. Although neutrophils are usually present in skin lesions of early syphilis, the role of these cells in T. pallidum infection has barely been investigated. Neutrophils are short-lived cells that undergo constitutive apoptosis, and phagocytosis usually accelerates this process. Here, we demonstrated that human polymorphonuclear neutrophils (hPMNs) could phagocytose T. pallidum in vitro. An unexpected discovery was that T. pallidum inhibited hPMNs apoptosis markedly in an opsonin-independent manner. Furthermore, this phenomenon was not affected by bacterial viability, as detected by annexin V, morphology studies, and TUNEL staining. Exploration of the underlying mechanism showed that expression of the cleaved forms of caspase-3, -8, and -9 and effector caspase activity were diminished significantly in T. pallidum-infected hPMNs. T. pallidum also impaired staurosporine- and anti-Fas-induced signaling for neutrophil apoptosis. Of note, these effects were accompanied by inducing the autocrine production of the anti-apoptotic cytokine IL-8. Taken together, our data revealed that T. pallidum could inhibit the apoptosis of hPMNs through intrinsic and extrinsic pathways and provide new insights for understanding the pathogenicity mechanisms of T. pallidum.
Collapse
Affiliation(s)
- Jianye Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Simin Lu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Kang Zheng
- Clinical Laboratory, Hengyang Central Hospital, Hengyang, China
| | - Zhangping He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Weiwei Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Jie Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Ningyuan Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Yafeng Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China; Department of Clinical Laboratory, the Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Dejun Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China
| | - Man Xu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China.
| | - Yimou Wu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical College, Institution of Pathogenic Biology, University of South China, Hengyang, China.
| |
Collapse
|
13
|
Kremserová S, Kocurková A, Chorvátová M, Klinke A, Kubala L. Myeloperoxidase Deficiency Alters the Process of the Regulated Cell Death of Polymorphonuclear Neutrophils. Front Immunol 2022; 13:707085. [PMID: 35211113 PMCID: PMC8860816 DOI: 10.3389/fimmu.2022.707085] [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: 05/08/2021] [Accepted: 01/18/2022] [Indexed: 01/17/2023] Open
Abstract
Polymorphonuclear neutrophils (PMNs) play a key role in host defense. However, their massive accumulation at the site of inflammation can delay regenerative healing processes and can initiate pathological inflammatory processes. Thus, the efficient clearance of PMNs mediated by the induction of regulated cell death is a key process preventing the development of these pathological conditions. Myeloperoxidase (MPO), a highly abundant enzyme in PMN granules, primarily connected with PMN defense machinery, is suggested to play a role in PMN-regulated cell death. However, the contribution of MPO to the mechanisms of PMN cell death remains incompletely characterized. Herein, the process of the cell death of mouse PMNs induced by three different stimuli – phorbol 12-myristate 13-acetate (PMA), opsonized streptococcus (OST), and N-formyl-met-leu-phe (fMLP) – was investigated. MPO-deficient PMNs revealed a significantly decreased rate of cell death characterized by phosphatidylserine surface exposure and cell membrane permeabilization. An inhibitor of MPO activity, 4-aminobenzoic acid hydrazide, did not exhibit a significant effect on PMA-induced cell death compared to MPO deficiency. Interestingly, only the limited activation of markers related to apoptotic cell death was observed (e.g. caspase 8 activation, Bax expression) and they mostly did not correspond to phosphatidylserine surface exposure. Furthermore, a marker characterizing autophagy, cleavage of LC3 protein, as well as histone H3 citrullination and its surface expression was observed. Collectively, the data show the ability of MPO to modulate the life span of PMNs primarily through the potentiation of cell membrane permeabilization and phosphatidylserine surface exposure.
Collapse
Affiliation(s)
- Silvie Kremserová
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia
| | - Anna Kocurková
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czechia
| | - Michaela Chorvátová
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Anna Klinke
- Clinic of General and Interventional Cardiology/Angiology, Agnes Wittenborg Institute of Translational Cardiovascular Research, Herz- und Diabeteszentrum NRW, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Lukáš Kubala
- Institute of Biophysics, Academy of Sciences of the Czech Republic, Brno, Czechia.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czechia
| |
Collapse
|
14
|
de los Reyes AA, Kim Y. Optimal regulation of tumour-associated neutrophils in cancer progression. ROYAL SOCIETY OPEN SCIENCE 2022; 9:210705. [PMID: 35127110 PMCID: PMC8808100 DOI: 10.1098/rsos.210705] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 11/19/2021] [Indexed: 06/14/2023]
Abstract
In a tumour microenvironment, tumour-associated neutrophils could display two opposing differential phenotypes: anti-tumour (N1) and pro-tumour (N2) effector cells. Converting N2 to N1 neutrophils provides innovative therapies for cancer treatment. In this study, a mathematical model for N1-N2 dynamics describing the cancer survival and immune inhibition in response to TGF-β and IFN-β is considered. The effects of exogenous intervention of TGF-β inhibitor and IFN-β are examined in order to enhance N1 recruitment to combat tumour progression. Our approach employs optimal control theory to determine drug infusion protocols that could minimize tumour volume with least administration cost possible. Four optimal control scenarios corresponding to different therapeutic strategies are explored, namely, TGF-β inhibitor control only, IFN-β control only, concomitant TGF-β inhibitor and IFN-β controls, and alternating TGF-β inhibitor and IFN-β controls. For each scheme, different initial conditions are varied to depict different pathophysiological condition of a cancer patient, leading to adaptive treatment schedule. TGF-β inhibitor and IFN-β drug dosages, total drug amount, infusion times and relative cost of drug administrations are obtained under various circumstances. The control strategies achieved could guide in designing individualized therapeutic protocols.
Collapse
Affiliation(s)
- Aurelio A. de los Reyes
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon 34126, Republic of Korea
- Institute of Mathematics, University of the Philippines Diliman, Quezon City 1101, Philippines
| | - Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul 05029, Republic of Korea
- Mathematical Biosciences Institute, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Belchamber KBR, Hughes MJ, Spittle DA, Walker EM, Sapey E. New Pharmacological Tools to Target Leukocyte Trafficking in Lung Disease. Front Immunol 2021; 12:704173. [PMID: 34367163 PMCID: PMC8334730 DOI: 10.3389/fimmu.2021.704173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 06/24/2021] [Indexed: 01/13/2023] Open
Abstract
Infection and inflammation of the lung results in the recruitment of non-resident immune cells, including neutrophils, eosinophils and monocytes. This swift response should ensure clearance of the threat and resolution of stimuli which drive inflammation. However, once the threat is subdued this influx of immune cells should be followed by clearance of recruited cells through apoptosis and subsequent efferocytosis, expectoration or retrograde migration back into the circulation. This cycle of cell recruitment, containment of threat and then clearance of immune cells and repair is held in exquisite balance to limit host damage. Advanced age is often associated with detrimental changes to the balance described above. Cellular functions are altered including a reduced ability to traffic accurately towards inflammation, a reduced ability to clear pathogens and sustained inflammation. These changes, seen with age, are heightened in lung disease, and most chronic and acute lung diseases are associated with an exaggerated influx of immune cells, such as neutrophils, to the airways as well as considerable inflammation. Indeed, across many lung diseases, pathogenesis and progression has been associated with the sustained presence of trafficking cells, with examples including chronic diseases such as Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis and acute infections such as Pneumonia and Pneumonitis. In these instances, there is evidence that dysfunctional and sustained recruitment of cells to the airways not only increases host damage but impairs the hosts ability to effectively respond to microbial invasion. Targeting leukocyte migration in these instances, to normalise cellular responses, has therapeutic promise. In this review we discuss the current evidence to support the trafficking cell as an immunotherapeutic target in lung disease, and which potential mechanisms or pathways have shown promise in early drug trials, with a focus on the neutrophil, as the quintessential trafficking immune cell.
Collapse
Affiliation(s)
- Kylie B. R. Belchamber
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Michael J. Hughes
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Daniella A. Spittle
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Eloise M. Walker
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
| | - Elizabeth Sapey
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, United Kingdom
- NIHR Clinical Research Facility Birmingham, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
16
|
Endothelial Dysfunction and Neutrophil Degranulation as Central Events in Sepsis Physiopathology. Int J Mol Sci 2021; 22:ijms22126272. [PMID: 34200950 PMCID: PMC8230689 DOI: 10.3390/ijms22126272] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022] Open
Abstract
Sepsis is a major health problem worldwide. It is a time-dependent disease, with a high rate of morbidity and mortality. In this sense, an early diagnosis is essential to reduce these rates. The progressive increase of both the incidence and prevalence of sepsis has translated into a significant socioeconomic burden for health systems. Currently, it is the leading cause of noncoronary mortality worldwide and represents one of the most prevalent pathologies both in hospital emergency services and in intensive care units. In this article, we review the role of both endothelial dysfunction and neutrophil dysregulation in the physiopathology of this disease. The lack of a key symptom in sepsis makes it difficult to obtain a quick and accurate diagnosis of this condition. Thus, it is essential to have fast and reliable diagnostic tools. In this sense, the use of biomarkers can be a very important alternative when it comes to achieving these goals. Both new biomarkers and treatments related to endothelial dysfunction and neutrophil dysregulation deserve to be further investigated in order to open new venues for the diagnosis, treatment and prognosis of sepsis.
Collapse
|
17
|
Pérez-Figueroa E, Álvarez-Carrasco P, Ortega E, Maldonado-Bernal C. Neutrophils: Many Ways to Die. Front Immunol 2021; 12:631821. [PMID: 33746968 PMCID: PMC7969520 DOI: 10.3389/fimmu.2021.631821] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 02/08/2021] [Indexed: 12/21/2022] Open
Abstract
Neutrophils or polymorphonuclear leukocytes (PMN) are key participants in the innate immune response for their ability to execute different effector functions. These cells express a vast array of membrane receptors that allow them to recognize and eliminate infectious agents effectively and respond appropriately to microenvironmental stimuli that regulate neutrophil functions, such as activation, migration, generation of reactive oxygen species, formation of neutrophil extracellular traps, and mediator secretion, among others. Currently, it has been realized that activated neutrophils can accomplish their effector functions and simultaneously activate mechanisms of cell death in response to different intracellular or extracellular factors. Although several studies have revealed similarities between the mechanisms of cell death of neutrophils and other cell types, neutrophils have distinctive properties, such as a high production of reactive oxygen species (ROS) and nitrogen species (RNS), that are important for their effector function in infections and pathologies such as cancer, autoimmune diseases, and immunodeficiencies, influencing their cell death mechanisms. The present work offers a synthesis of the conditions and molecules implicated in the regulation and activation of the processes of neutrophil death: apoptosis, autophagy, pyroptosis, necroptosis, NETosis, and necrosis. This information allows to understand the duality encountered by PMNs upon activation. The effector functions are carried out to eliminate invading pathogens, but in several instances, these functions involve activation of signaling cascades that culminate in the death of the neutrophil. This process guarantees the correct elimination of pathogenic agents, damaged or senescent cells, and the timely resolution of the inflammation that is essential for the maintenance of homeostasis in the organism. In addition, they alert the organism when the immunological system is being deregulated, promoting the activation of other cells of the immune system, such as B and T lymphocytes, which produce cytokines that potentiate the microbicide functions.
Collapse
Affiliation(s)
- Erandi Pérez-Figueroa
- Unidad de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City, Mexico
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Pablo Álvarez-Carrasco
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Enrique Ortega
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Carmen Maldonado-Bernal
- Unidad de Investigación en Inmunología y Proteómica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City, Mexico
| |
Collapse
|
18
|
Ledo C, Gonzalez CD, Garofalo A, Sabbione F, Keitelman IA, Giai C, Stella I, Trevani AS, Gómez MI. Protein A Modulates Neutrophil and Keratinocyte Signaling and Survival in Response to Staphylococcus aureus. Front Immunol 2021; 11:524180. [PMID: 33692774 PMCID: PMC7937904 DOI: 10.3389/fimmu.2020.524180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 12/29/2020] [Indexed: 01/18/2023] Open
Abstract
The type 1 TNF-α receptor (TNFR1) has a central role in initiating both pro-inflammatory and pro-apoptotic signaling cascades in neutrophils. Considering that TNFR1 signals Staphylococcus aureus protein A (SpA), the aim of this study was to explore the interaction of this bacterial surface protein with neutrophils and keratinocytes to underscore the signaling pathways that may determine the fate of these innate immune cells in the infected tissue during staphylococcal skin infections. Using human neutrophils cultured in vitro and isogenic staphylococcal strains expressing or not protein A, we demonstrated that SpA is a potent inducer of IL-8 in neutrophils and that the induction of this chemokine is dependent on the SpA-TNFR1 interaction and p38 activation. In addition to IL-8, protein A induced the expression of TNF-α and MIP-1α highlighting the importance of SpA in the amplification of the inflammatory response. Protein A contributed to reduce neutrophil mortality prolonging their lifespan upon the encounter with S. aureus. Signaling initiated by SpA modulated the type of neutrophil cell death in vitro and during skin and soft tissue infections (SSTI) in vivo triggering the apoptotic pathway instead of necrosis. Moreover, SpA induced pro-inflammatory cytokines in keratinocytes, modulating their survival in vitro and preventing the exacerbated necrosis and ulceration of the epithelium during SSTI in vivo. Taken together, these results highlight the importance of the inflammatory signaling induced by protein A in neutrophils and skin epithelial cells. The ability of protein A to modulate the neutrophil/epithelial cell death program in the skin is of clinical relevance considering that lysis of neutrophils and epithelial cells will promote an intense inflammatory response and contribute to tissue damage, a non-desirable feature of complicated SSTI.
Collapse
Affiliation(s)
- Camila Ledo
- Centro de Estudios Biomédicos, Aplicados y Desarrollo (CEBBAD), Departamento de Ciencias Biológicas y Biomédicas, Universidad Maimonides, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Cintia D Gonzalez
- Instituto de investigaciones en Microbiología y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ailin Garofalo
- Instituto de investigaciones en Microbiología y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Florencia Sabbione
- Departamento de Inmunología, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Irene A Keitelman
- Departamento de Inmunología, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Constanza Giai
- Instituto de investigaciones en Microbiología y Parasitología Médica (IMPaM), Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Inés Stella
- Facultad de Ciencias de la Salud, Universidad Maimónides, Buenos Aires, Argentina
| | - Analía S Trevani
- Departamento de Inmunología, Instituto de Medicina Experimental (IMEX)-CONICET, Academia Nacional de Medicina, Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marisa I Gómez
- Centro de Estudios Biomédicos, Aplicados y Desarrollo (CEBBAD), Departamento de Ciencias Biológicas y Biomédicas, Universidad Maimonides, Buenos Aires, Argentina.,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Departamento de Microbiología, Parasitología e Inmunología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
19
|
Orosz A, Walzog B, Mócsai A. In Vivo Functions of Mouse Neutrophils Derived from HoxB8-Transduced Conditionally Immortalized Myeloid Progenitors. THE JOURNAL OF IMMUNOLOGY 2020; 206:432-445. [PMID: 33310871 DOI: 10.4049/jimmunol.2000807] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/06/2020] [Indexed: 12/31/2022]
Abstract
Although neutrophils play important roles in immunity and inflammation, their analysis is strongly hindered by their short-lived and terminally differentiated nature. Prior studies reported conditional immortalization of myeloid progenitors using retroviral expression of an estrogen-dependent fusion protein of the HoxB8 transcription factor. This approach allowed the long-term culture of mouse myeloid progenitors (HoxB8 progenitors) in estrogen-containing media, followed by differentiation toward neutrophils upon estrogen withdrawal. Although several reports confirmed the in vitro functional responsiveness of the resulting differentiated cells (HoxB8 neutrophils), little is known about their capacity to perform in vivo neutrophil functions. We have addressed this issue by an in vivo transplantation approach. In vitro-generated HoxB8 neutrophils showed a neutrophil-like phenotype and were able to perform conventional neutrophil functions, like respiratory burst, chemotaxis, and phagocytosis. The i.v. injection of HoxB8 progenitors into lethally irradiated recipients resulted in the appearance of circulating donor-derived HoxB8 neutrophils. In vivo-differentiated HoxB8 neutrophils were able to migrate to the inflamed peritoneum and to phagocytose heat-killed Candida particles. The reverse passive Arthus reaction could be induced in HoxB8 chimeras but not in irradiated, nontransplanted control animals. Repeated injection of HoxB8 progenitors also allowed us to maintain stable circulating HoxB8 neutrophil counts for several days. Injection of arthritogenic K/B×N serum triggered robust arthritis in HoxB8 chimeras, but not in irradiated, nontransplanted control mice. Taken together, our results indicate that HoxB8 progenitor-derived neutrophils are capable of performing various in vivo neutrophil functions, providing a framework for using the HoxB8 system for the in vivo analysis of neutrophil function.
Collapse
Affiliation(s)
- Anita Orosz
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary
| | - Barbara Walzog
- Department of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig-Maximilians University of Munich, 82152 Planegg-Martinsried, Germany; and.,Walter Brendel Centre of Experimental Medicine, University Hospital, Ludwig-Maximilians University of Munich, 81377 Munich, Germany
| | - Attila Mócsai
- Department of Physiology, Semmelweis University School of Medicine, 1094 Budapest, Hungary;
| |
Collapse
|
20
|
Current knowledge into the role of the peptidylarginine deiminase (PAD) enzyme family in cardiovascular disease. Eur J Pharmacol 2020; 891:173765. [PMID: 33249073 DOI: 10.1016/j.ejphar.2020.173765] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/12/2020] [Accepted: 11/23/2020] [Indexed: 11/20/2022]
Abstract
Peptidylarginine deiminase (PAD) family members have a vital role in maintaining the stability of the extracellular matrix (ECM) during remodelling in several heart diseases. PAD-mediated deamination, or citrullination, has been studied in different physiological and pathological conditions in the body. However, the role of PAD isoforms has not been fully studied in cardiovascular system. Citrullination is a post-translational modification that involves conversion of peptidyl-based arginine to peptidyl-based citrulline by PAD family members in a calcium-dependent manner. Upregulation of PADs have been observed in various cardiovascular diseases, including venous thrombosis, cardiac fibrosis, heart failure, atherosclerosis, coronary heart disease and acute inflammation. In this review, experimental aspects of in vivo and in vitro studies related to the roles PAD isoforms in cardiovascular diseases including mechanisms, pathophysiological and therapeutic properties are discussed. Pharmacological strategies for targeting PAD family proteins in cardiac diseases have not yet been studied. Furthermore, the role played by PAD family members in the remodelling process during the progression of cardiovascular diseases is not fully understood.
Collapse
|
21
|
Wang LT, Wang HH, Chiang HC, Huang LY, Chiu SK, Siu LK, Liu KJ, Yen ML, Yen BL. Human Placental MSC-Secreted IL-1β Enhances Neutrophil Bactericidal Functions during Hypervirulent Klebsiella Infection. Cell Rep 2020; 32:108188. [PMID: 32997996 DOI: 10.1016/j.celrep.2020.108188] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 12/06/2019] [Accepted: 09/02/2020] [Indexed: 12/29/2022] Open
Abstract
Hypervirulent Klebsiella pneumoniae (hvKP) causes severe infections even in healthy individuals by escaping surveillance and killing from polymorphonuclear neutrophils (PMNs), the first-line leukocytes in bacterial infections; moreover, the emergence of multidrug-resistant strains further limits treatment options. We therefore assess whether multilineage mesenchymal stem cells (MSCs), best known for immunomodulation toward T cells, could be therapeutic for highly virulent bacterial infections via modulation of PMNs. We find that both bone marrow MSCs and placental MSCs (PMSCs) preserve in vitro PMN survival, but only PMSCs significantly enhance multiple PMN bactericidal functions, including phagocytosis, through secretion of interleukin-1β (IL-1β). PMSC treatment of hvKP-infected mice suppresses T and natural killer (NK) cell responses as expected but can preferentially recruit PMNs and enhance antibacterial functions to allow for disease survival; IL-1β knockdown in PMSCs significantly decreases hvKP clearance, worsening survival and resulting in 100% lethality. Our data strongly implicate the possible use of PMSCs for infections of PMN-resistant hvKP strains.
Collapse
Affiliation(s)
- Li-Tzu Wang
- Department of Obstetrics and Gynecology, National Taiwan University (NTU) Hospital and College of Medicine, NTU, Taipei 100, Taiwan
| | - Hsiu-Huan Wang
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| | - Hui-Chun Chiang
- Department of Obstetrics and Gynecology, National Taiwan University (NTU) Hospital and College of Medicine, NTU, Taipei 100, Taiwan
| | - Li-Yueh Huang
- National Institute of Infectious Diseases and Vaccinology, NHRI, Zhunan 350, Taiwan
| | - Sheng-Kang Chiu
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - L Kristopher Siu
- National Institute of Infectious Diseases and Vaccinology, NHRI, Zhunan 350, Taiwan; Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan; Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, NHRI, Zhunan 350, Taiwan; Institute of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung University, Tainan 701, Taiwan; School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei 110, Taiwan
| | - Men-Luh Yen
- Department of Obstetrics and Gynecology, National Taiwan University (NTU) Hospital and College of Medicine, NTU, Taipei 100, Taiwan.
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan; Department of Obstetrics and Gynecology, Cathay General Hospital Shiji, New Taipei 221, Taiwan.
| |
Collapse
|
22
|
Identification of key protein-coding genes and lncRNAs in spontaneous neutrophil apoptosis. Sci Rep 2019; 9:15106. [PMID: 31641174 PMCID: PMC6805912 DOI: 10.1038/s41598-019-51597-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 10/04/2019] [Indexed: 01/17/2023] Open
Abstract
Polymorphonuclear leukocytes (PMNs) are the most abundant cells of the innate immune system in humans, and spontaneous PMN apoptosis plays crucial roles in maintaining neutrophil homeostasis and resolving inflammation. However, the detailed mechanisms of spontaneous PMN apoptosis remain to be elucidated. By analysis of the public microarray dataset GSE37416, we identified a total of 3050 mRNAs and 220 long non-coding RNAs (lncRNAs) specifically expressed during PMN apoptosis in a time-dependent manner. By short time-series expression miner (STEM) analysis, Gene Ontology analysis, and lncRNA-mRNA co-expression network analyses, we identified some key molecules specifically related to PMN apoptosis. STEM analysis identified 12 gene profiles with statistically significance, including 2 associated with apoptosis. Protein-protein interaction (PPI) network analysis of the genes from 2 profiles and lncRNA-mRNA co-expression network analysis identified a 12-gene hub (including NFκB1 and BIRC3) associated with apoptosis, as well as 2 highly correlated lncRNAs (THAP9-AS1, and AL021707.6). We experimentally examined the expression profiles of two mRNA (NFκB1 and BIRC3) and two lncRNAs (THAP9-AS1 andAL021707.6) by quantitative real-time polymerase chain reaction to confirm their time-dependent expressions. These data altogether demonstrated that these genes are involved in the regulation of spontaneous neutrophil apoptosis and the corresponding gene products could also serve as potential key regulatory molecules for PMN apoptosis and/or therapeutic targets for over-reactive inflammatory response caused by the abnormality in PMN apoptosis.
Collapse
|
23
|
Paparini DE, Choudhury RH, Vota DM, Karolczak-Bayatti M, Finn-Sell S, Grasso EN, Hauk VC, Ramhorst R, Pérez Leirós C, Aplin JD. Vasoactive intestinal peptide shapes first-trimester placenta trophoblast, vascular, and immune cell cooperation. Br J Pharmacol 2019; 176:964-980. [PMID: 30726565 DOI: 10.1111/bph.14609] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 12/10/2018] [Accepted: 01/01/2019] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND AND PURPOSE Extravillous trophoblast (EVT) cells are responsible for decidual stromal invasion, vascular transformation, and the recruitment and functional modulation of maternal leukocytes in the first-trimester pregnant uterus. An early disruption of EVT function leads to placental insufficiency underlying pregnancy complications such as preeclampsia and fetal growth restriction. Vasoactive intestinal peptide (VIP) is a vasodilating and immune modulatory factor synthesized by trophoblast cells. However, its role in first-trimester placenta has not been explored. Here, we tested the hypothesis that VIP is involved in first-trimester EVT outgrowth, spiral artery remodelling, balancing angiogenesis, and maintenance of immune homeostasis. EXPERIMENTAL APPROACH First-trimester placental tissue (five to nine weeks of gestation) was collected, and was used for EVT outgrowth experiments, immunofluorescence, isolation of decidual natural killer (dNK) cells and decidual macrophages (dMA), and functional assays. Peripheral blood monocytes were differentiated with GM-CSF and used for angiogenesis assays. KEY RESULTS In decidua basalis, VIP+ EVT were observed sprouting from cell columns and lining spiral arterioles. EVT migrating from placental explants were also VIP+. VIP increased EVT outgrowth and IL-10 release, whereas it decreased pro-inflammatory cytokine production in EVT, dNK cells, and dMA. VIP disrupted endothelial cell networks, both directly and indirectly via an effect on macrophages. CONCLUSION AND IMPLICATIONS The results suggest that VIP assists the progress of EVT invasion and vessel remodelling in first-trimester placental bed in an immunologically "silent" milieu. The effects of VIP in the present ex vivo human placental model endorse its potential as a therapeutic candidate for deep placentation disorders.
Collapse
Affiliation(s)
- Daniel E Paparini
- IQUIBICEN-CONICET, School of Sciences, University of Buenos Aires, Buenos Aires, Argentina.,Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, UK
| | - Ruhul H Choudhury
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, UK
| | - Daiana M Vota
- IQUIBICEN-CONICET, School of Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Magdalena Karolczak-Bayatti
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, UK
| | - Sarah Finn-Sell
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, UK
| | - Esteban N Grasso
- IQUIBICEN-CONICET, School of Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Vanesa C Hauk
- IQUIBICEN-CONICET, School of Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Rosanna Ramhorst
- IQUIBICEN-CONICET, School of Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- IQUIBICEN-CONICET, School of Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - John D Aplin
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester, UK
| |
Collapse
|
24
|
Kim Y, Lee D, Lee J, Lee S, Lawler S. Role of tumor-associated neutrophils in regulation of tumor growth in lung cancer development: A mathematical model. PLoS One 2019; 14:e0211041. [PMID: 30689655 PMCID: PMC6349324 DOI: 10.1371/journal.pone.0211041] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023] Open
Abstract
Neutrophils display rapid and potent innate immune responses in various diseases. Tumor-associated neutrophils (TANs) however either induce or overcome immunosuppressive functions of the tumor microenvironment through complex tumor-stroma crosstalk. We developed a mathematical model to address the question of how phenotypic alterations between tumor suppressive N1 TANS, and tumor promoting N2 TANs affect nonlinear tumor growth in a complex tumor microenvironment. The model provides a visual display of the complex behavior of populations of TANs and tumors in response to various TGF-β and IFN-β stimuli. In addition, the effect of anti-tumor drug administration is incorporated in the model in an effort to achieve optimal anti-tumor efficacy. The simulation results from the mathematical model were in good agreement with experimental data. We found that the N2-to-N1 ratio (N21R) index is positively correlated with aggressive tumor growth, suggesting that this may be a good prognostic factor. We also found that the antitumor efficacy increases when the relative ratio (Dap) of delayed apoptotic cell death of N1 and N2 TANs is either very small or relatively large, providing a basis for therapeutically targeting prometastatic N2 TANs.
Collapse
Affiliation(s)
- Yangjin Kim
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
- Mathematical Biosciences Institute, Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| | - Donggu Lee
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| | - Junho Lee
- Department of Mathematics, Konkuk University, Seoul, Republic of Korea
| | - Seongwon Lee
- Division of Mathematical Models, National Institute for Mathematical Sciences, Daejeon, Republic of Korea
| | - Sean Lawler
- Department of neurosurgery, Harvard Medical School & Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| |
Collapse
|
25
|
Lokwani R, Wark PAB, Baines KJ, Barker D, Simpson JL. Hypersegmented airway neutrophils and its association with reduced lung function in adults with obstructive airway disease: an exploratory study. BMJ Open 2019; 9:e024330. [PMID: 30696679 PMCID: PMC6352776 DOI: 10.1136/bmjopen-2018-024330] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES The significance of neutrophilic inflammation in obstructive airway disease remains controversial. Recent studies have demonstrated presence of an active neutrophil population in systemic circulation, featuring hypersegmented morphology, with high oxidative burst and functional plasticity in inflammatory conditions. The aim of this study was to characterise neutrophil subsets in bronchial lavage (BL) of obstructive airway disease participants (asthma, chronic obstructive pulmonary disease (COPD) and bronchiectasis) and healthy controls on the basis of nuclear morphology and to assess the association between neutrophil subsets and the clinical parameters of the obstructive airway disease participants. DESIGN A cross-sectional exploratory study. SETTING John Hunter Hospital and Hunter Medical Research Institute, Australia. PARTICIPANTS Seventy-eight adults with obstructive airway disease comprised those with stable asthma (n=39), COPD (n=20) and bronchiectasis (n=19) and 20 healthy controls. MATERIALS AND METHODS Cytospins were prepared and neutrophil subsets were classified based on nuclear morphology into hypersegmented (>4 lobes), normal (2-4 lobes) and banded (1 lobe) neutrophils and enumerated. RESULTS Neutrophils from each subset were identified in all participants. Numbers of hypersegmented neutrophils were elevated in participants with airway disease compared with healthy controls (p<0.001). Both the number and the proportion of hypersegmented neutrophils were highest in COPD participants (median (Q1-Q3) of 1073.6 (258.8-2742) × 102/mL and 24.5 (14.0-46.5)%, respectively). An increased proportion of hypersegmented neutrophils in airway disease participants was significantly associated with lower forced expiratory volume in 1 s/forced vital capacity per cent (Spearman's r=-0.322, p=0.004). CONCLUSION Neutrophil heterogeneity is common in BL and is associated with more severe airflow obstruction in adults with airway disease. Further work is required to elucidate the functional consequences of hypersegmented neutrophils in the pathogenesis of disease.
Collapse
Affiliation(s)
- Ravi Lokwani
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, New Lambton, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, New Lambton, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton, New South Wales, Australia
| | - Katherine J Baines
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, New Lambton, New South Wales, Australia
| | - Daniel Barker
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, New Lambton, New South Wales, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, New South Wales, Australia
- Faculty of Health and Medicine, School of Medicine and Public Health, University of Newcastle, New Lambton, New South Wales, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, New Lambton, New South Wales, Australia
| |
Collapse
|
26
|
Dong Y, Lagarde J, Xicota L, Corne H, Chantran Y, Chaigneau T, Crestani B, Bottlaender M, Potier MC, Aucouturier P, Dorothée G, Sarazin M, Elbim C. Neutrophil hyperactivation correlates with Alzheimer's disease progression. Ann Neurol 2019; 83:387-405. [PMID: 29369398 DOI: 10.1002/ana.25159] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 01/23/2018] [Accepted: 01/23/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Recent studies have underlined the effect of systemic inflammation on the pathophysiology of Alzheimer's disease (AD). Neutrophils are key components of early innate immunity and contribute to uncontrolled systemic inflammation if not tightly regulated. The aim of our study was to fully characterize human circulating neutrophils at different disease stages in AD. METHODS We analyzed neutrophil phenotypes and functions in 42 patients with AD (16 with mild cognitive impairment and 26 with dementia), and compared them to 22 age-matched healthy subjects. This study was performed directly in whole blood to avoid issues with data interpretation related to cell isolation procedures. RESULTS Blood samples from AD patients with dementia revealed neutrophil hyperactivation associated with increased reactive oxygen species production and increased levels of intravascular neutrophil extravascular traps. The homeostasis of circulating neutrophils in these patients also changed: The ratio between the harmful hyperreactive CXCR4high /CD62Llow senescent and the CD16bright /CD62Ldim immunosuppressive neutrophil subsets rose in the later stage of the disease. These abnormalities were greater in fast-decliner than in slow-decliner patients. INTERPRETATION Our results indicate that the inflammatory properties of circulating neutrophils shift as the percentage of aged neutrophils expands in patients with AD-changes that may play an instrumental role in establishing systemic chronic inflammation. Most important, our data strongly suggest that the neutrophil phenotype may be associated with the rate of cognitive decline and may thus constitute an innovative and prognostic blood biomarker in patients with AD. Ann Neurol 2018;83:387-405.
Collapse
Affiliation(s)
- Yuan Dong
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine, team "Immune System, Neuroinflammation and Neurodegenerative Diseases", Hôpital Saint-Antoine, Paris, France
| | - Julien Lagarde
- Unit of Neurology of Memory and Language, Centre Hospitalier Sainte Anne, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Laboratoire Imagerie Moléculaire In Vivo (IMIV), UMR 1023 Inserm/CEA/Université Paris Sud-ERL 9218 CNRS; CEA/I2BM/Service Hospitalier Frédéric Joliot, Orsay, France
| | - Laura Xicota
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U1127, CNRS UMR 7225, UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Hélène Corne
- Unit of Neurology of Memory and Language, Centre Hospitalier Sainte Anne, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Yannick Chantran
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine, team "Immune System, Neuroinflammation and Neurodegenerative Diseases", Hôpital Saint-Antoine, Paris, France
| | - Thomas Chaigneau
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine, team "Immune System, Neuroinflammation and Neurodegenerative Diseases", Hôpital Saint-Antoine, Paris, France
| | - Bruno Crestani
- APHP, Hôpital Bichat, Service de Pneumologie A, Centre de référence des maladies pulmonaires rares, Paris, France
| | - Michel Bottlaender
- Laboratoire Imagerie Moléculaire In Vivo (IMIV), UMR 1023 Inserm/CEA/Université Paris Sud-ERL 9218 CNRS; CEA/I2BM/Service Hospitalier Frédéric Joliot, Orsay, France.,UNIACT, NeuroSpin, Institut d'Imagerie Biomédicale, Direction des sciences du vivant, Commissariat à I'Energie Atomique, Gif-sur-Yvette, France
| | - Marie-Claude Potier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM U1127, CNRS UMR 7225, UMRS 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Pierre Aucouturier
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine, team "Immune System, Neuroinflammation and Neurodegenerative Diseases", Hôpital Saint-Antoine, Paris, France
| | - Guillaume Dorothée
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine, team "Immune System, Neuroinflammation and Neurodegenerative Diseases", Hôpital Saint-Antoine, Paris, France
| | - Marie Sarazin
- Unit of Neurology of Memory and Language, Centre Hospitalier Sainte Anne, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Laboratoire Imagerie Moléculaire In Vivo (IMIV), UMR 1023 Inserm/CEA/Université Paris Sud-ERL 9218 CNRS; CEA/I2BM/Service Hospitalier Frédéric Joliot, Orsay, France
| | - Carole Elbim
- Sorbonne Universités, UPMC Univ Paris 06, INSERM, Centre de Recherche Saint-Antoine, team "Immune System, Neuroinflammation and Neurodegenerative Diseases", Hôpital Saint-Antoine, Paris, France
| |
Collapse
|
27
|
Moraes JA, Frony AC, Barcellos-de-Souza P, Menezes da Cunha M, Brasil Barbosa Calcia T, Benjamim CF, Boisson-Vidal C, Barja-Fidalgo C. Downregulation of Microparticle Release and Pro-Inflammatory Properties of Activated Human Polymorphonuclear Neutrophils by LMW Fucoidan. J Innate Immun 2018; 11:330-346. [PMID: 30557873 PMCID: PMC6738154 DOI: 10.1159/000494220] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 09/21/2018] [Accepted: 09/21/2018] [Indexed: 12/16/2022] Open
Abstract
Exposition of neutrophils (polymorphonuclear neutrophils, PMNs) to bacterial products triggers exacerbated activation of these cells, increasing their harmful effects on host tissues. We evaluated the possibility of interfering with the classic immune innate responses of human PMNs exposed to bacterial endotoxin (lipopolysaccharide, LPS), and further stimulated with bacterial formyl peptide (N-formyl-methionine-leucine-phenylalanine, fMLP). We showed that the low- molecular-weight fucoidan (LMW-Fuc), a polysaccharide extracted from brown algae, attenuated the exacerbated activation induced by fMLP on LPS-primed PMNs, in vitro, impairing chemotaxis, NET formation, and the pro-survival and pro-oxidative effects. LMW-Fuc also inhibited the activation of canonical signaling pathways, AKT, bad, p47phox and MLC, activated by the exposition of PMN to bacterial products. The activation of PMN by sequential exposure to LPS and fMLP induced the release of L-selectin+ microparticles, which were able to trigger extracellular reactive oxygen species production by fresh PMNs and macrophages. Furthermore, we observed that LMW-Fuc inhibited microparticle release from activated PMN. In vivo experiments showed that circulating PMN-derived microparticles could be detected in mice exposed to bacterial products (LPS/fMLP), being downregulated in animals treated with LMW-Fuc. The data highlight the autocrine and paracrine role of pro-inflammatory microparticles derived from activated PMN and demonstrate the anti-inflammatory effects of LMW-Fuc on these cells.
Collapse
Affiliation(s)
- João Alfredo Moraes
- Laboratório de Farmacologia Celular e Molecular, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Biologia RedOx, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Clara Frony
- Laboratório de Farmacologia Celular e Molecular, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Barcellos-de-Souza
- Laboratório de Farmacologia Celular e Molecular, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcel Menezes da Cunha
- Núcleo Multidisciplinar de Pesquisa em Biologia-NUMPEX-BIO, Universidade Federal do Rio de Janeiro, Xerém, Brazil
| | | | - Claudia Farias Benjamim
- Laboratório de Imunologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Christina Barja-Fidalgo
- Laboratório de Farmacologia Celular e Molecular, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil,
| |
Collapse
|
28
|
Regulation of neutrophils in type 2 immune responses. Curr Opin Immunol 2018; 54:115-122. [PMID: 30015087 DOI: 10.1016/j.coi.2018.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/14/2018] [Accepted: 06/27/2018] [Indexed: 01/09/2023]
Abstract
Type 2 immune responses contribute to the resistance to helminths and toxins as well as several physiological processes. Although they usually do not participate in type 2 immune responses, neutrophils have been shown in mice to enhance the anti-helminth response, but they also contribute to increased target tissue damage. Increased pathology and morbidity is also observed in type 2 immune-mediated disorders, such as allergic asthma, when neutrophils become a predominant subset of the infiltrate. How neutrophil recruitment is regulated during type 2 immune responses is now starting to become clear, with recent data showing that signaling via the prototypic type 2 cytokine interleukin-4 receptor mediates direct and indirect inhibitory actions on neutrophils in mice and humans.
Collapse
|
29
|
Jie H, He Y, Huang X, Zhou Q, Han Y, Li X, Bai Y, Sun E. Necrostatin-1 enhances the resolution of inflammation by specifically inducing neutrophil apoptosis. Oncotarget 2017; 7:19367-81. [PMID: 27027357 PMCID: PMC4991389 DOI: 10.18632/oncotarget.8346] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 03/08/2016] [Indexed: 12/20/2022] Open
Abstract
Neutrophils play a central role in innate immunity and are rapidly recruited to sites of infection and injury. Neutrophil apoptosis is essential for the successful resolution of inflammation. Necrostatin-1 (Nec-1,methyl-thiohydantoin-tryptophan (MTH-Trp)), is a potent and specific inhibitor of necroptosis[1] (a newly identified type of cell death representing a form of programmed necrosis or regulated non apoptotic cell death) by inhibiting the receptor interacting protein 1(RIP1) kinase. Here we report that Nec-1 specifically induces caspase-dependent neutrophils apoptosis and overrides powerful anti-apoptosis signaling from survival factors such as GM-CSF and LPS. We showed that Nec-1 markedly enhanced the resolution of established neutrophil-dependent inflammation in LPS-induced acute lung injury in mice. We also provided evidence that Nec-1 promoted apoptosis by reducing the expression of the anti-apoptotic protein Mcl-1 and increasing the expression of pro-apoptotic protein Bax. Thus, Nec-1 is not only an inhibitor of necroptosis, but also a promoter of apoptosis, of neutrophils, enhancing the resolution of established inflammation by inducing apoptosis of inflammatory cells. Our results suggest that Nec-1 may have potential roles for the treatment of diseases with increased or persistent inflammatory responses.
Collapse
Affiliation(s)
- Hongyu Jie
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Institute of Clinical Immunology, Academy of Orthopedics, Guangzhou, Guangdong, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Institute of Clinical Immunology, Academy of Orthopedics, Guangzhou, Guangdong, China
| | - Xuechan Huang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Institute of Clinical Immunology, Academy of Orthopedics, Guangzhou, Guangdong, China
| | - Qingyou Zhou
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Institute of Clinical Immunology, Academy of Orthopedics, Guangzhou, Guangdong, China
| | - Yanping Han
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Institute of Clinical Immunology, Academy of Orthopedics, Guangzhou, Guangdong, China.,Hospital of South China Normal University, Guangzhou, Guangdong, China
| | - Xing Li
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Institute of Clinical Immunology, Academy of Orthopedics, Guangzhou, Guangdong, China
| | - Yongkun Bai
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Institute of Clinical Immunology, Academy of Orthopedics, Guangzhou, Guangdong, China
| | - Erwei Sun
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China.,Institute of Clinical Immunology, Academy of Orthopedics, Guangzhou, Guangdong, China
| |
Collapse
|
30
|
Lin J, He K, Zhao G, Li C, Hu L, Zhu G, Niu Y, Hao G. Mincle inhibits neutrophils and macrophages apoptosis in A. fumigatus keratitis. Int Immunopharmacol 2017; 52:101-109. [DOI: 10.1016/j.intimp.2017.08.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 06/30/2017] [Accepted: 08/09/2017] [Indexed: 11/16/2022]
|
31
|
Abstract
The engulfment of apoptotic cells by phagocytes, a process referred to as efferocytosis, is essential for maintenance of normal tissue homeostasis and a prerequisite for the resolution of inflammation. Neutrophils are the predominant circulating white blood cell in humans, and contain an arsenal of toxic substances that kill and degrade microbes. Neutrophils are short-lived and spontaneously die by apoptosis. This review will highlight how the engulfment of apoptotic neutrophils by human phagocytes occurs, how heterogeneity of phagocyte populations influences efferocytosis signaling, and downstream consequences of efferocytosis. The efferocytosis of apoptotic neutrophils by macrophages promotes anti-inflammatory signaling, prevents neutrophil lysis, and dampens immune responses. Given the immunomodulatory properties of efferocytosis, understanding pathways that regulate and enhance efferocytosis could be harnessed to combat infection and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Mallary C Greenlee-Wacker
- Inflammation Program, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Veterans Administration Medical Center, Iowa City, IA, USA
| |
Collapse
|
32
|
Kinkead LC, Fayram DC, Allen LAH. Francisella novicida inhibits spontaneous apoptosis and extends human neutrophil lifespan. J Leukoc Biol 2017; 102:815-828. [PMID: 28550119 DOI: 10.1189/jlb.4ma0117-014r] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 01/09/2023] Open
Abstract
Francisella novicida is a Gram-negative bacterium that is closely related to the highly virulent facultative intracellular pathogen, Francisella tularensis Data published by us and others demonstrate that F. tularensis virulence correlates directly with its ability to impair constitutive apoptosis and extend human neutrophil lifespan. In contrast, F. novicida is attenuated in humans, and the mechanisms that account for this are incompletely defined. Our published data demonstrate that F. novicida binds natural IgG that is present in normal human serum, which in turn, elicits NADPH oxidase activation that does not occur in response to F. tularensis As it is established that phagocytosis and oxidant production markedly accelerate neutrophil death, we predicted that F. novicida may influence the neutrophil lifespan in an opsonin-dependent manner. To test this hypothesis, we quantified bacterial uptake, phosphatidylserine (PS) externalization, and changes in nuclear morphology, as well as the kinetics of procaspase-3, -8, and -9 processing and activation. To our surprise, we discovered that F. novicida not only failed to accelerate neutrophil death but also diminished and delayed apoptosis in a dose-dependent, but opsonin-independent, manner. In keeping with this, studies of conditioned media (CM) showed that neutrophil longevity could be uncoupled from phagocytosis and that F. novicida stimulated neutrophil secretion of CXCL8. Taken together, the results of this study reveal shared and unique aspects of the mechanisms used by Francisella species to manipulate neutrophil lifespan and as such, advance understanding of cell death regulation during infection.
Collapse
Affiliation(s)
- Lauren C Kinkead
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.,Iowa City VA Medical Center, Iowa City, Iowa, USA
| | - Drew C Fayram
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA.,Department of Microbiology, University of Iowa, Iowa City, Iowa, USA
| | - Lee-Ann H Allen
- Inflammation Program, University of Iowa, Iowa City, Iowa, USA; .,Department of Microbiology, University of Iowa, Iowa City, Iowa, USA.,Iowa City VA Medical Center, Iowa City, Iowa, USA.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA; and
| |
Collapse
|
33
|
Moris D, Schizas D, Karaolanis G, Karavokyros I. eComment: Neutrophil-to-lymphocyte ratio: Ambitious but Ambiguous marker. Interact Cardiovasc Thorac Surg 2017; 24:559. [PMID: 28375464 DOI: 10.1093/icvts/ivx052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Demetrios Moris
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Dimitrios Schizas
- Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Karaolanis
- Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Karavokyros
- Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
34
|
Ghrenassia E, Guihot A, Dong Y, Robinet P, Fontaine T, Lacombe K, Lescot T, Meyohas MC, Elbim C. First Report of CD4 Lymphopenia and Defective Neutrophil Functions in a Patient with Amebiasis Associated with CMV Reactivation and Severe Bacterial and Fungal Infections. Front Microbiol 2017; 8:203. [PMID: 28243230 PMCID: PMC5303735 DOI: 10.3389/fmicb.2017.00203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/27/2017] [Indexed: 11/20/2022] Open
Abstract
We report the case of a patient with acute necrotizing colitis due to invasive amebiasis associated with CD4 lymphopenia and impaired neutrophil responses. The course of the disease was characterized by CMV reactivation and severe and recurrent bacterial and fungal infections, which might be related to the decreased CD4 T cell count and the impaired functional capacities of neutrophils, respectively. The clinical outcome was positive with normalization of both CD4 cell count and neutrophil functions.
Collapse
Affiliation(s)
- Etienne Ghrenassia
- AP-HP, Hôpital Saint-Antoine, Service des Maladies Infectieuses et Tropicales Paris, France
| | - Amélie Guihot
- Département d'Immunologie, AP-HP, Hôpital Pitié-SalpêtrièreParis, France; DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Universités, UPMC Univ Paris 06Paris, France; Institut National de la Santé et de la Recherche Médicale, U1135, Centre d'Immunologie et des Maladies Infectieuses-ParisParis, France
| | - Yuan Dong
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche Saint-Antoine, UMR-S 938 Paris, France
| | - Pauline Robinet
- DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Universités, UPMC Univ Paris 06Paris, France; Institut National de la Santé et de la Recherche Médicale, U1135, Centre d'Immunologie et des Maladies Infectieuses-ParisParis, France
| | | | - Karine Lacombe
- AP-HP, Hôpital Saint-Antoine, Service des Maladies Infectieuses et Tropicales Paris, France
| | - Thomas Lescot
- Unité de Réanimation Chirurgicale Digestive, Département D'anesthésie et de Réanimation Chirurgicale, AP-HP, Hôpital Saint-Antoine Paris, France
| | - Marie-Caroline Meyohas
- AP-HP, Hôpital Saint-Antoine, Service des Maladies Infectieuses et Tropicales Paris, France
| | - Carole Elbim
- DHU FAST, CR7, Centre d'Immunologie et des Maladies Infectieuses, Sorbonne Universités, UPMC Univ Paris 06Paris, France; Institut National de la Santé et de la Recherche Médicale, U1135, Centre d'Immunologie et des Maladies Infectieuses-ParisParis, France
| |
Collapse
|
35
|
Role of granule proteases in the life and death of neutrophils. Biochem Biophys Res Commun 2017; 482:473-481. [PMID: 28212734 DOI: 10.1016/j.bbrc.2016.11.086] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/08/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023]
Abstract
Neutrophils constitute a crucial component of the innate immune defenses against microbes. Produced in the bone marrow and patrolling in blood vessels, neutrophils are recruited to injured tissues and are immediately active to contain pathogen invasion. Neutrophils undergo programmed cell death by multiple, context-specific pathways, which have consequences on immunopathology and disease outcome. Studies in the last decade indicate additional functions for neutrophils - or a subset of neutrophils - in modulating adaptive responses and tumor progression. Neutrophil granules contain abundant amounts of various proteases, which are directly implicated in protective and pathogenic functions of neutrophils. It now emerges that neutral serine proteases such as cathepsin G and proteinase-3 also contribute to the neutrophil life cycle, but do so via different pathways than that of the aspartate protease cathepsin D and that of mutants of the serine protease elastase. The aim of this review is to appraise the present knowledge of the function of neutrophil granule proteases and their inhibitors in neutrophil cell death, and to integrate these findings in the current understandings of neutrophil life cycle and programmed cell death pathways.
Collapse
|
36
|
Calo G, Sabbione F, Vota D, Paparini D, Ramhorst R, Trevani A, Pérez Leirós C. Trophoblast cells inhibit neutrophil extracellular trap formation and enhance apoptosis through vasoactive intestinal peptide-mediated pathways. Hum Reprod 2016; 32:55-64. [PMID: 27932441 DOI: 10.1093/humrep/dew292] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/28/2016] [Accepted: 10/14/2016] [Indexed: 12/24/2022] Open
Abstract
STUDY QUESTION Do human trophoblast cells modulate neutrophil extracellular trap (NET) formation, reactive oxygen species (ROS) synthesis and neutrophil apoptosis through mechanisms involving vasoactive intestinal peptide (VIP)? SUMMARY ANSWER Trophoblast cells inhibited NET formation and ROS synthesis and enhanced neutrophil apoptosis through VIP-mediated pathways in a model of maternal-placental interaction. WHAT IS KNOWN ALREADY Immune homeostasis maintenance at the maternal-placental interface is mostly coordinated by trophoblast cells. Neutrophil activation and NET formation increases in pregnancies complicated by exacerbated pro-inflammatory responses. VIP has anti-inflammatory and immunosuppressant effects and is synthesized by trophoblast cells. STUDY DESIGN, SIZE, DURATION This is a laboratory-based observational study that sampled circulating neutrophils from 50 healthy volunteers to explore their response in vitro to factors derived from human trophoblast cells. PARTICIPANTS/MATERIALS, SETTING, METHODS Peripheral blood neutrophils were isolated from healthy volunteers and tested in vitro with first trimester trophoblast cell line (Swan-71 and HTR8) conditioned media (CM) or with VIP. The effect of VIP and trophoblast CM on NET formation was assessed by co-localization of elastase and DNA by confocal microscopy, DNA release and elastase activity measurement. Neutrophil apoptosis was determined by flow cytometry or fluorescence microscopy. ROS formation was assessed by flow cytometry with a fluorescent probe. VIP silencing was performed by siRNA transfection. For phagocytosis of apoptotic neutrophils, autologous monocytes were sampled, and engulfment and cytokines were assessed by flow cytometry and ELISA. MAIN RESULTS AND THE ROLE OF CHANCE Trophoblast CM and 10 nM VIP promoted neutrophil deactivation by preventing phorbol myristate acetate-induced NET formation and ROS synthesis while they increased neutrophil spontaneous apoptosis and reversed the anti-apoptotic effect of lipopolysaccharide (all P < 0.05 versus control). The effects of trophoblast CM were prevented by a VIP antagonist or when VIP knocked-down trophoblast cells were used (P < 0.05 versus control). Neutrophils driven to apoptosis by trophoblast CM could be rapidly engulfed by monocytes without increasing IL-12 production. LARGE SCALE DATA Not applicable. LIMITATIONS, REASONS FOR CAUTION The mechanisms of neutrophil deactivation by trophoblast VIP are based on the results obtained with neutrophils drawn from peripheral blood of healthy individuals interacting with trophoblast cell lines in vitro. These studies were designed to investigate biological processes at the cellular and molecular level; therefore, they have the limitations of studies in vitro and it is not possible to ascertain if these mechanisms operate similarly in vivo. We tested 50 neutrophil samples from healthy volunteers that have a normal variability in their responses. Cell lines derived from human trophoblast were used, and we cannot rule out a differential behavior of trophoblast cells in contact with neutrophils in vivo. WIDER IMPLICATIONS OF THE FINDINGS Results presented here are consistent with an active mechanism through which neutrophils in contact with trophoblast cells would be deactivated and silently cleared by decidual macrophages throughout pregnancy. They support a novel immunomodulatory role of trophoblast VIP on neutrophils at the placenta, providing new clues for pharmacological targeting of immune and trophoblast cells in pregnancy complications associated with exacerbated inflammation. STUDY FUNDING/COMPETING INTERESTS This work was funded by the National Agency of Sciences and Technology (PICT 2011-0144, 2014-0657 and 2013-2177) and University of Buenos Aires (UBACyT 20020130100040BA, 20020150100161BA and 20020130100744BA). The authors declare no competing interests.
Collapse
Affiliation(s)
- Guillermina Calo
- CONICET, Facultad de Ciencias Exactas y Naturales, Instituto de Química Biológica (IQUIBICEN), Universidad de Buenos Aires, Ciudad Universitaria, Pab II. Fourth Floor, 1428 Buenos Aires, Argentina
| | - Florencia Sabbione
- CONICET, Facultad de Medicina, Instituto de Medicina Experimental, Academia Nacional de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Daiana Vota
- CONICET, Facultad de Ciencias Exactas y Naturales, Instituto de Química Biológica (IQUIBICEN), Universidad de Buenos Aires, Ciudad Universitaria, Pab II. Fourth Floor, 1428 Buenos Aires, Argentina
| | - Daniel Paparini
- CONICET, Facultad de Ciencias Exactas y Naturales, Instituto de Química Biológica (IQUIBICEN), Universidad de Buenos Aires, Ciudad Universitaria, Pab II. Fourth Floor, 1428 Buenos Aires, Argentina
| | - Rosanna Ramhorst
- CONICET, Facultad de Ciencias Exactas y Naturales, Instituto de Química Biológica (IQUIBICEN), Universidad de Buenos Aires, Ciudad Universitaria, Pab II. Fourth Floor, 1428 Buenos Aires, Argentina
| | - Analía Trevani
- CONICET, Facultad de Medicina, Instituto de Medicina Experimental, Academia Nacional de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- CONICET, Facultad de Ciencias Exactas y Naturales, Instituto de Química Biológica (IQUIBICEN), Universidad de Buenos Aires, Ciudad Universitaria, Pab II. Fourth Floor, 1428 Buenos Aires, Argentina
| |
Collapse
|
37
|
Martinez EC, Garg R, Shrivastava P, Gomis S, van Drunen Littel-van den Hurk S. Intranasal treatment with a novel immunomodulator mediates innate immune protection against lethal pneumonia virus of mice. Antiviral Res 2016; 135:108-119. [PMID: 27771388 PMCID: PMC7126411 DOI: 10.1016/j.antiviral.2016.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/07/2016] [Accepted: 10/18/2016] [Indexed: 12/26/2022]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections in infants and young children. There are no licensed RSV vaccines available, and the few treatment options for high-risk individuals are either extremely costly or cause severe side effects and toxicity. Immunomodulation mediated by a novel formulation consisting of the toll-like receptor 3 agonist poly(I:C), an innate defense regulator peptide and a polyphosphazene (P-I-P) was evaluated in the context of lethal infection with pneumonia virus of mice (PVM). Intranasal delivery of a single dose of P-I-P protected adult mice against PVM when given 24 h prior to challenge. These animals experienced minimal weight loss, no clinical disease, 100% survival, and reduced lung pathology. Similar clinical outcomes were observed in mice treated up to 3 days prior to infection. P-I-P pre-treatment induced early mRNA and protein expression of key chemokine and cytokine genes, reduced the recruitment of neutrophils and eosinophils, decreased virus titers in the lungs, and modulated the delayed exacerbated nature of PVM disease without any short-term side effects. On day 14 post-infection, P-I-P-treated mice were confirmed to be PVM-free. These results demonstrate the capacity of this formulation to prevent PVM and possibly other viral respiratory infections. P-I-P pre-treatment, consisting of poly(I:C), IDR peptide and PCEP, was tested in the context of PVM infection in mice. P-I-P confers complete protection against lethal PVM infection by reducing clinical signs and immunopathology. P-I-P minimizes viral titers in the lungs reduces the influx of neutrophils and eosinophils into the tissue. P-I-P induces early upregulation of genes involved in host defense without any observable adverse effects. Survivor mice were PVM negative, suggesting that P-I-P mediates the successfully clearance of the virus in vivo.
Collapse
Affiliation(s)
- Elisa C Martinez
- Department of Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, 107 Wiggins Road, S7N 5E5, Canada; Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Ravendra Garg
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Pratima Shrivastava
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada
| | - Susantha Gomis
- Department of Veterinary Pathology, Western College of Veterinary Medicine (WCVM), University of Saskatchewan, Saskatoon, Saskatchewan, 52 Campus Drive, S7N 5B4, Canada
| | - Sylvia van Drunen Littel-van den Hurk
- Department of Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, 107 Wiggins Road, S7N 5E5, Canada; Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, Saskatchewan, 120 Veterinary Road, S7N 5E3, Canada.
| |
Collapse
|
38
|
Huang P, Zhou Y, Liu Z, Zhang P. Interaction between ANXA1 and GATA-3 in Immunosuppression of CD4 + T Cells. Mediators Inflamm 2016; 2016:1701059. [PMID: 27833268 PMCID: PMC5090097 DOI: 10.1155/2016/1701059] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/20/2016] [Indexed: 01/29/2023] Open
Abstract
Decreased Th1/Th2 ratio is one of the major characteristics of immunosuppression in sepsis. Both membrane adhesive protein Annexin-A1 (ANXA1) and transcription factor GATA-3 have been reported to play important roles in T cell differentiation. However, the relationship between ANXA1 and GATA-3 in Th1/Th2 shift is unknown. Our study investigated the interaction effects of ANXA1 and GATA-3 to influence T cell differentiation in CD4+ T cells. We found that GATA-3 and ANXA1 were coexpressed on Th0/Th1/Th2 cytoplasm and nuclear. Overexpressed ANXA1 significantly increased the expression of IFNγ and reduced IL-4 expression in T cells, while ANXA1-silenced T cells exhibited decreased production of IFNγ and increased production of IL-4. Knockdown of ANXA1 promoted higher expression level of GATA-3 and low level of T-box transcription factor (T-bet/Tbx21). Further study demonstrated that ANXA1 regulated GATA-3 expression through the formyl peptide receptor like-1 (FPRL-1) downstream signaling pathways ERK and PKB/Akt. These results suggested that ANXA1 modulates GATA-3/T-bet expression induced Th0/Th1 differentiation. Moreover, we found that GATA-3 inhibited ANXA1 expression by binding to its promoter for the first time. It is proposed that the interactions between ANXA1 and GATA-3 may provide clues to understand the immunosuppression and have potential as new therapeutic targets in immunotherapy after sepsis.
Collapse
Affiliation(s)
- Peng Huang
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuxiang Zhou
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zan Liu
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pihong Zhang
- Department of Burns and Reconstructive Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
39
|
Malachowa N, Kobayashi SD, Quinn MT, DeLeo FR. NET Confusion. Front Immunol 2016; 7:259. [PMID: 27446089 PMCID: PMC4923183 DOI: 10.3389/fimmu.2016.00259] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 06/17/2016] [Indexed: 12/17/2022] Open
Abstract
Neutrophils are arguably the most important white blood cell for defense against bacterial and fungal infections. These leukocytes are produced in high numbers on a daily basis in humans and are recruited rapidly to injured/infected tissues. Phagocytosis and subsequent intraphagosomal killing and digestion of microbes have historically been the accepted means by which neutrophils carry out their role in innate host defense. Indeed, neutrophils contain and produce numerous cytotoxic molecules, including antimicrobial peptides, proteases, and reactive oxygen species, that are highly effective at killing the vast majority of ingested microbes. On the other hand, it is these characteristics - high numbers and toxicity - that endow neutrophils with the potential to injure and destroy host tissues. This potential is borne out by many inflammatory processes and diseases. Therefore, it is not surprising that host mechanisms exist to control virtually all steps in the neutrophil activation process and to prevent unintended neutrophil activation and/or lysis during the resolution of inflammatory responses or during steady-state turnover. The notion that neutrophil extracellular traps (NETs) form by cytolysis as a standard host defense mechanism seems inconsistent with these aforementioned neutrophil "containment" processes. It is with this caveat in mind that we provide perspective on the role of NETs in human host defense and disease.
Collapse
Affiliation(s)
- Natalia Malachowa
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Hamilton, MT , USA
| | - Scott D Kobayashi
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Hamilton, MT , USA
| | - Mark T Quinn
- Department of Microbiology and Immunology, Montana State University , Bozeman, MT , USA
| | - Frank R DeLeo
- Laboratory of Bacteriology, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Hamilton, MT , USA
| |
Collapse
|
40
|
Vasoactive Intestinal Peptide modulates trophoblast-derived cell line function and interaction with phagocytic cells through autocrine pathways. Sci Rep 2016; 6:26364. [PMID: 27212399 PMCID: PMC4876379 DOI: 10.1038/srep26364] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 04/27/2016] [Indexed: 02/08/2023] Open
Abstract
Trophoblast cells migrate and invade the decidual stroma in a tightly regulated process to maintain immune homeostasis at the maternal-placental interface during the first weeks of pregnancy. Locally synthesized factors modulate trophoblast cell function and their interaction with maternal leukocytes to promote the silent clearance of apoptotic cells. The vasoactive intestinal peptide (VIP) is a pleiotropic polypeptide with trophic and anti-inflammatory effects in murine pregnancy models. We explored the effect of VIP on two human first trimester trophoblast cell lines, particularly on their migration, invasiveness and interaction with phagocytic cells, and the signalling and regulatory pathways involved. We found that VIP enhanced trophoblast cell migration and invasion through the activation of high affinity VPAC receptors and PKA-CRE signalling pathways. VIP knocked-down trophoblast cells showed reduced migration in basal and leukemic inhibitor factor (LIF)-elicited conditions. In parallel, VIP-silenced trophoblast cells failed to induce the phagocytosis of apoptotic bodies and the expression of immunosuppressant markers by human monocytes. Our results suggest that VIP-mediated autocrine pathways regulate trophoblast cell function and contribute to immune homeostasis maintenance at placentation and may provide new clues for therapeutic intervention in pregnancies complicated by defective deep placentation.
Collapse
|
41
|
Bermejo-Martin JF, Andaluz-Ojeda D, Almansa R, Gandía F, Gómez-Herreras JI, Gomez-Sanchez E, Heredia-Rodríguez M, Eiros JM, Kelvin DJ, Tamayo E. Defining immunological dysfunction in sepsis: A requisite tool for precision medicine. J Infect 2016; 72:525-36. [PMID: 26850357 DOI: 10.1016/j.jinf.2016.01.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 01/24/2016] [Accepted: 01/26/2016] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Immunological dysregulation is now recognised as a major pathogenic event in sepsis. Stimulation of immune response and immuno-modulation are emerging approaches for the treatment of this disease. Defining the underlying immunological alterations in sepsis is important for the design of future therapies with immuno-modulatory drugs. METHODS Clinical studies evaluating the immunological response in adult patients with Sepsis and published in PubMed were reviewed to identify features of immunological dysfunction. For this study we used key words related with innate and adaptive immunity. RESULTS Ten major features of immunological dysfunction (FID) were identified involving quantitative and qualitative alterations of [antigen presentation](FID1), [T and B lymphocytes] (FID2), [natural killer cells] (FID3), [relative increase in T regulatory cells] (FID4), [increased expression of PD-1 and PD-ligand1](FID5), [low levels of immunoglobulins](FID6), [low circulating counts of neutrophils and/or increased immature forms in non survivors](FID7), [hyper-cytokinemia] (FID8), [complement consumption] (FID9), [defective bacterial killing by neutrophil extracellular traps](FID10). CONCLUSIONS This review article identified ten major features associated with immunosuppression and immunological dysregulation in sepsis. Assessment of these features could help in utilizing precision medicine for the treatment of sepsis with immuno-modulatory drugs.
Collapse
Affiliation(s)
- Jesús F Bermejo-Martin
- Infection and Immunity Medical Investigation Unit (IMI), Hospital Clínico Universitario de Valladolid, SACYL/IECSCYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - David Andaluz-Ojeda
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Medicina Intensiva, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Raquel Almansa
- Infection and Immunity Medical Investigation Unit (IMI), Hospital Clínico Universitario de Valladolid, SACYL/IECSCYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Francisco Gandía
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Medicina Intensiva, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Jose Ignacio Gómez-Herreras
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología y Reanimación, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Esther Gomez-Sanchez
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología y Reanimación, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - María Heredia-Rodríguez
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología y Reanimación, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Jose Maria Eiros
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - David J Kelvin
- Division of Experimental Therapeutics, Toronto General Hospital Research Institute, University Health Network, 200 Elizabeth Street, Toronto, ON M5G 2C4, Canada; Sezione di Microbiologia Sperimentale e Clinica, Dipartimento di Scienze Biomediche, Universita' degli Studi di Sassari, Piazza Università, 21, 07100 Sassari SS, Italy; International Institute of Infection and Immunity, Shantou University Medical College, 22 Xinling Road, Shantou, 515041 Guangdong Province, PR China.
| | - Eduardo Tamayo
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología y Reanimación, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| |
Collapse
|
42
|
Sauce D, Dong Y, Campillo-Gimenez L, Casulli S, Bayard C, Autran B, Boddaert J, Appay V, Elbim C. Reduced Oxidative Burst by Primed Neutrophils in the Elderly Individuals Is Associated With Increased Levels of the CD16bright/CD62LdimImmunosuppressive Subset. J Gerontol A Biol Sci Med Sci 2016; 72:163-172. [DOI: 10.1093/gerona/glw062] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 03/16/2016] [Indexed: 12/28/2022] Open
|
43
|
Charles TP, Shellito JE. Human Immunodeficiency Virus Infection and Host Defense in the Lungs. Semin Respir Crit Care Med 2016; 37:147-56. [PMID: 26974294 DOI: 10.1055/s-0036-1572553] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immunosuppression associated with human immunodeficiency virus (HIV) infection impacts all components of host defense against pulmonary infection. Cells within the lung have altered immune function and are important reservoirs for HIV infection. The host immune response to infected lung cells further compromises responses to a secondary pathogenic insult. In the upper respiratory tract, mucociliary function is impaired and there are decreased levels of salivary immunoglobulin A. Host defenses in the lower respiratory tract are controlled by alveolar macrophages, lymphocytes, and polymorphonuclear leukocytes. As HIV infection progresses, lung CD4 T cells are reduced in number causing a lack of activation signals from CD4 T cells and impaired defense by macrophages. CD8 T cells, on the other hand, are increased in number and cause lymphocytic alveolitis. Specific antibody responses by B-lymphocytes are decreased and opsonization of microorganisms is impaired. These observed defects in host defense of the respiratory tract explain the susceptibility of HIV-infected persons for oropharyngeal candidiasis, bacterial pneumonia, Pneumocystis pneumonia, and other opportunistic infections.
Collapse
Affiliation(s)
- Tysheena P Charles
- Section of Pulmonary/Critical Care & Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Judd E Shellito
- Section of Pulmonary/Critical Care & Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
44
|
McCracken JM, Kinkead LC, McCaffrey RL, Allen LAH. Francisella tularensis Modulates a Distinct Subset of Regulatory Factors and Sustains Mitochondrial Integrity to Impair Human Neutrophil Apoptosis. J Innate Immun 2016; 8:299-313. [PMID: 26906922 DOI: 10.1159/000443882] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/09/2016] [Indexed: 12/13/2022] Open
Abstract
Tularemia is a disease characterized by profound neutrophil accumulation and tissue destruction. The causative organism, Francisella tularensis, is a facultative intracellular bacterium that replicates in neutrophil cytosol, inhibits caspase activation and profoundly prolongs cell lifespan. Here, we identify unique features of this infection and provide fundamental insight into the mechanisms of apoptosis inhibition. Mitochondria are critical regulators of neutrophil apoptosis. We demonstrate that F. tularensis significantly inhibits Bax translocation and Bid processing during 24-48 h of infection, and in this manner sustains mitochondrial integrity. Downstream of mitochondria, X-linked inhibitor of apoptosis protein (XIAP) and proliferating cell nuclear antigen (PCNA) inhibit caspase-9 and caspase-3 by direct binding. Notably, we find that PCNA disappeared rapidly and selectively from infected cells, thereby demonstrating that it is not essential for neutrophil survival, whereas upregulation of calpastatin correlated with diminished calpain activity and reduced XIAP degradation. In addition, R-roscovitine is a cyclin-dependent kinase inhibitor developed for the treatment of cancer; it also induces neutrophil apoptosis and can promote the resolution of several infectious and inflammatory disorders. We confirm the ability of R-roscovitine to induce neutrophil apoptosis, but also demonstrate that its efficacy is significantly impaired by F. tularensis. Collectively, our findings advance the understanding of neutrophil apoptosis and its capacity to be manipulated by pathogenic bacteria.
Collapse
Affiliation(s)
- Jenna M McCracken
- Inflammation Program, University of Iowa and VA Medical Center, Iowa City, Iowa, USA
| | | | | | | |
Collapse
|
45
|
Wayne EC, Chandrasekaran S, Mitchell MJ, Chan MF, Lee RE, Schaffer CB, King MR. TRAIL-coated leukocytes that prevent the bloodborne metastasis of prostate cancer. J Control Release 2015; 223:215-223. [PMID: 26732555 DOI: 10.1016/j.jconrel.2015.12.048] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 12/22/2015] [Accepted: 12/23/2015] [Indexed: 12/24/2022]
Abstract
Prostate cancer, once it has progressed from its local to metastatic form, is a disease with poor prognosis and limited treatment options. Here we demonstrate an approach using nanoscale liposomes conjugated with E-selectin adhesion protein and Apo2L/TRAIL (TNF-related apoptosis-inducing ligand) apoptosis ligand that attach to the surface of leukocytes and rapidly clear viable cancer cells from circulating blood in the living mouse. For the first time, it is shown that such an approach can be used to prevent the spontaneous formation and growth of metastatic tumors in an orthotopic xenograft model of prostate cancer, by greatly reducing the number of circulating tumor cells. We conclude that the use of circulating leukocytes as a carrier for the anti-cancer protein TRAIL could be an effective tool to directly target circulating tumor cells for the prevention of prostate cancer metastasis, and potentially other cancers that spread through the bloodstream.
Collapse
Affiliation(s)
- Elizabeth C Wayne
- Meinig School of Biomedical Engineering, Cornell University, 526 N. Campus Rd, Weill Hall, Ithaca, NY 14853, United States
| | - Siddarth Chandrasekaran
- Meinig School of Biomedical Engineering, Cornell University, 526 N. Campus Rd, Weill Hall, Ithaca, NY 14853, United States
| | - Michael J Mitchell
- Meinig School of Biomedical Engineering, Cornell University, 526 N. Campus Rd, Weill Hall, Ithaca, NY 14853, United States
| | - Maxine F Chan
- Meinig School of Biomedical Engineering, Cornell University, 526 N. Campus Rd, Weill Hall, Ithaca, NY 14853, United States
| | - Rachel E Lee
- Meinig School of Biomedical Engineering, Cornell University, 526 N. Campus Rd, Weill Hall, Ithaca, NY 14853, United States
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, 526 N. Campus Rd, Weill Hall, Ithaca, NY 14853, United States
| | - Michael R King
- Meinig School of Biomedical Engineering, Cornell University, 526 N. Campus Rd, Weill Hall, Ithaca, NY 14853, United States.
| |
Collapse
|
46
|
Distinct Functions of Neutrophil in Cancer and Its Regulation. Mediators Inflamm 2015; 2015:701067. [PMID: 26648665 PMCID: PMC4663337 DOI: 10.1155/2015/701067] [Citation(s) in RCA: 134] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/27/2015] [Indexed: 01/13/2023] Open
Abstract
Neutrophils are the most abundant of all white blood cells in the human circulation and are usually associated with inflammation and with fighting infections. In recent years the role immune cells play in cancer has been a matter of increasing interest. In this context the function of neutrophils is controversial as neutrophils were shown to possess both tumor promoting and tumor limiting properties. Here we provide an up-to-date review of the pro- and antitumor properties neutrophils possess as well as the environmental cues that regulate these distinct functions.
Collapse
|
47
|
Espinasse MA, Pépin A, Virault-Rocroy P, Szely N, Chollet-Martin S, Pallardy M, Biola-Vidamment A. Glucocorticoid-Induced Leucine Zipper Is Expressed in Human Neutrophils and Promotes Apoptosis through Mcl-1 Down-Regulation. J Innate Immun 2015; 8:81-96. [PMID: 26384220 DOI: 10.1159/000439052] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/29/2015] [Indexed: 01/19/2023] Open
Abstract
Glucocorticoid-induced leucine zipper (GILZ) is a potent anti-inflammatory protein, the expression of which is mainly induced by glucocorticoids (GCs) in haematopoietic cells. GILZ regulates signal transduction pathways of inflammation and plays a role in cell survival. The objective of this study was to evaluate the expression and mechanisms of action of GILZ in the apoptosis of human neutrophils. GILZ expression was induced by GCs in human neutrophils, enhanced upon phosphatidylinositol 3-kinase inhibition and resulted in apoptosis amplification. We then stably transfected PLB-985 cells with the human gilz gene and differentiated both control and GILZ-overexpressing clones in neutrophil-like cells. GILZ overexpression in PLB-985 cells led to an exacerbated apoptosis, associated with caspase-3, caspase-9 and caspase-8 activations, and a loss of mitochondrial potential, suggesting that GILZ-induced apoptosis used the mitochondrial pathway. The expression of BH3 interacting domain death agonist, Bcl-2 interacting mediator of cell death, annexin-A1 and Bcl-2-associated X was not affected in PLB-985-GILZ clones, but phosphorylation and subsequent proteasomal degradation of myeloid cell leukemia-1 (Mcl-1) were observed. Noteworthy, Mcl-1 phosphorylation was related to a significant and sustained activation of c-Jun N-terminal kinase (JNK) in PLB-985-GILZ clones. These results reveal GILZ to be a new actor in apoptosis regulation in neutrophil-like cells involving JNK and Mcl-1.
Collapse
Affiliation(s)
- Marie-Alix Espinasse
- UMR996 - Inflammation, Chemokines and Immunopathology, Inserm, Universitx00E9; Paris-Sud, Universitx00E9; Paris-Saclay, Chx00E2;tenay-Malabry, France
| | | | | | | | | | | | | |
Collapse
|
48
|
Almansa R, Heredia-Rodríguez M, Gomez-Sanchez E, Andaluz-Ojeda D, Iglesias V, Rico L, Ortega A, Gomez-Pesquera E, Liu P, Aragón M, Eiros JM, Jiménez-Sousa MÁ, Resino S, Gómez-Herreras I, Bermejo-Martín JF, Tamayo E. Transcriptomic correlates of organ failure extent in sepsis. J Infect 2014; 70:445-56. [PMID: 25557485 DOI: 10.1016/j.jinf.2014.12.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVES Sepsis is characterised by the frequent presence of organ failure and marked immunologic alterations. We studied the association between the extent of organ failure and the transcriptomic response of septic patients. METHODS Gene expression profiles in the blood of 74 surgical patients with sepsis were compared with those of 30 surgical patients with no sepsis. Differentially expressed genes were assessed for their correlation with the sequential organ failure (SOFA) score. RESULTS The expression levels of a group of genes participating in the cell cycle (HIST1H1C, CKS2, CCNA2, CDK1, CCNB2, CIT, CCNB1, AURKA, RAD51), neutrophil protease activity (ELANE, ADORA3, MPO, MMP8, CTSG), IL-1R and IL-18R response correlated directly with SOFA and mortality. Genes involved in T cell (LCK, CD3G, CD3D, ZAP70, ICOS, CD3E, CD28, IL2RB, CD8B, CD8A, CD40LG, IL23A, CCL5, SH2D1A, ITK, CD247, TBX21, GATA3, CCR7, LEF1, STAT4) and NK cell immunity (CD244, KLRK1, KLRD1) were inversely associated with SOFA and mortality. CONCLUSIONS The extent of organ failure in sepsis correlates directly with the existence of imbalanced innate and adaptive responses at the transcriptomic level. Quantification of the expression levels of the genes identified here could contribute to the simultaneous assessment of disease severity and immunological alterations in sepsis.
Collapse
Affiliation(s)
- Raquel Almansa
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Unidad Apoyo a la Investigación, Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - María Heredia-Rodríguez
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Esther Gomez-Sanchez
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - David Andaluz-Ojeda
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Medicina Intensiva, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Verónica Iglesias
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Unidad Apoyo a la Investigación, Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Lucia Rico
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Unidad Apoyo a la Investigación, Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Alicia Ortega
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Unidad Apoyo a la Investigación, Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Estefanía Gomez-Pesquera
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Pilar Liu
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Marta Aragón
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Jose Maria Eiros
- Dpto de Microbiología, Universidad de Valladolid, Avenida Ramón y Cajal, 7, 47005 Valladolid, Spain.
| | - Maria Ángeles Jiménez-Sousa
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología - Instituto de Salud Carlos III, Carretera Majadahonda- Pozuelo km. 2, Majadahonda, 28220 Madrid, Spain.
| | - Salvador Resino
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Unidad de Infección Viral e Inmunidad, Centro Nacional de Microbiología - Instituto de Salud Carlos III, Carretera Majadahonda- Pozuelo km. 2, Majadahonda, 28220 Madrid, Spain.
| | - Ignacio Gómez-Herreras
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Jesús F Bermejo-Martín
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Unidad Apoyo a la Investigación, Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| | - Eduardo Tamayo
- Grupo de Investigación Biomédica en Cuidados Críticos (BioCritic), Hospital Clínico Universitario de Valladolid, SACYL/IECSYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain; Servicio de Anestesiología, Hospital Clínico Universitario de Valladolid, SACYL, Avenida Ramón y Cajal, 3, 47005 Valladolid, Spain.
| |
Collapse
|
49
|
Affiliation(s)
- J Charles Jennette
- Department of Pathology and Laboratory Medicine and University of North Carolina Kidney Center, University of North Carolina, Chapel Hill, North Carolina
| | - Ronald J Falk
- Department of Pathology and Laboratory Medicine and University of North Carolina Kidney Center, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
50
|
Campillo-Gimenez L, Casulli S, Dudoit Y, Seang S, Carcelain G, Lambert-Niclot S, Appay V, Autran B, Tubiana R, Elbim C. Neutrophils in antiretroviral therapy-controlled HIV demonstrate hyperactivation associated with a specific IL-17/IL-22 environment. J Allergy Clin Immunol 2014; 134:1142-52.e5. [PMID: 25042982 DOI: 10.1016/j.jaci.2014.05.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 05/30/2014] [Accepted: 05/30/2014] [Indexed: 12/22/2022]
Abstract
BACKGROUND Despite control of HIV infection under antiretroviral therapy (ART), immune T-cell activation persists in patients with controlled HIV infection, who are at higher risk of inflammatory diseases than the general population. PMNs play a key role in host defenses against invading microorganisms but also potentiate inflammatory reactions in cases of excessive or misdirected responses. OBJECTIVE The aim of our study was to analyze PMN functions in 60 ART-treated and controlled HIV-infected patients (viral load, <20 RNA copies/mL; CD4 count, ≥ 350 cells/mm(3)) with (HIV[I] group) and without (HIV[NI] group) diseases related to an inflammatory process and to compare them with 22 healthy control subjects. METHODS Flow cytometry was used to evaluate PMN functions in whole-blood conditions. We studied in parallel the activation markers of T lymphocytes and monocytes and the proinflammatory cytokine environment. RESULTS Blood samples from HIV-infected patients revealed basal PMN hyperactivation associated with deregulation of the apoptosis/necrosis equilibrium. Interestingly, this hyperactivation was greater in HIV(I) than HIV(NI) patients and contrasted with a lack of monocyte activation in both groups. The percentage of circulating cells producing IL-17 was also significantly higher in HIV-infected patients than in control subjects and was positively correlated with markers of basal PMN activation. In addition, the detection of IL-22 overproduction in HIV(NI) patients suggests that it might contribute to counteracting chronic inflammatory processes during HIV infection. CONCLUSIONS This study thus demonstrates the presence of highly activated PMNs in HIV-infected patients receiving effective ART and the association of these cells with a specific IL-17/IL-22 environment.
Collapse
Affiliation(s)
- Laure Campillo-Gimenez
- Sorbonne University, UPMC University Paris 06, Paris, France; INSERM, Centre d'Immunologie et des Maladies Infectieuses, UMR-S CR7, INSERM U1135, Paris, France
| | - Sarah Casulli
- Sorbonne University, UPMC University Paris 06, Paris, France; INSERM, Centre d'Immunologie et des Maladies Infectieuses, UMR-S CR7, INSERM U1135, Paris, France
| | - Yasmine Dudoit
- AP-HP, Hôpital Pitié-Salpêtrière, Service des maladies infectieuses et tropicales, Paris, France; Sorbonne University, UPMC University Paris 06, UMR-S 1136, Pierre Louis Institute of Epidemiology and Public Health, Paris, France
| | - Sophie Seang
- AP-HP, Hôpital Pitié-Salpêtrière, Service des maladies infectieuses et tropicales, Paris, France; Sorbonne University, UPMC University Paris 06, UMR-S 1136, Pierre Louis Institute of Epidemiology and Public Health, Paris, France
| | - Guislaine Carcelain
- INSERM, Centre d'Immunologie et des Maladies Infectieuses, UMR-S CR7, INSERM U1135, Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Laboratoire d'Immunologie Cellulaire et Tissulaire, Paris, France
| | - Sidonie Lambert-Niclot
- Sorbonne University, UPMC University Paris 06, UMR-S 1136, Pierre Louis Institute of Epidemiology and Public Health, Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Laboratoire de Virologie, Paris, France
| | - Victor Appay
- INSERM, Centre d'Immunologie et des Maladies Infectieuses, UMR-S CR7, INSERM U1135, Paris, France
| | - Brigitte Autran
- INSERM, Centre d'Immunologie et des Maladies Infectieuses, UMR-S CR7, INSERM U1135, Paris, France; AP-HP, Hôpital Pitié-Salpêtrière, Laboratoire d'Immunologie Cellulaire et Tissulaire, Paris, France
| | - Roland Tubiana
- AP-HP, Hôpital Pitié-Salpêtrière, Service des maladies infectieuses et tropicales, Paris, France; Sorbonne University, UPMC University Paris 06, UMR-S 1136, Pierre Louis Institute of Epidemiology and Public Health, Paris, France
| | - Carole Elbim
- Sorbonne University, UPMC University Paris 06, Paris, France; INSERM, Centre d'Immunologie et des Maladies Infectieuses, UMR-S CR7, INSERM U1135, Paris, France.
| |
Collapse
|