1
|
Tu H, Ren H, Jiang J, Shao C, Shi Y, Li P. Dying to Defend: Neutrophil Death Pathways and their Implications in Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306457. [PMID: 38044275 PMCID: PMC10885667 DOI: 10.1002/advs.202306457] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/06/2023] [Indexed: 12/05/2023]
Abstract
Neutrophils, accounting for ≈70% of human peripheral leukocytes, are key cells countering bacterial and fungal infections. Neutrophil homeostasis involves a balance between cell maturation, migration, aging, and eventual death. Neutrophils undergo different death pathways depending on their interactions with microbes and external environmental cues. Neutrophil death has significant physiological implications and leads to distinct immunological outcomes. This review discusses the multifarious neutrophil death pathways, including apoptosis, NETosis, pyroptosis, necroptosis, and ferroptosis, and outlines their effects on immune responses and disease progression. Understanding the multifaceted aspects of neutrophil death, the intersections among signaling pathways and ramifications of immunity will help facilitate the development of novel therapeutic methods.
Collapse
Affiliation(s)
- Haiyue Tu
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Haoyu Ren
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Junjie Jiang
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Changshun Shao
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Yufang Shi
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| | - Peishan Li
- The First Affiliated Hospital of Soochow UniversityState Key Laboratory of Radiation Medicine and ProtectionInstitutes for Translational MedicineSuzhou Medical College of Soochow UniversitySuzhouJiangsu215123China
| |
Collapse
|
2
|
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: 0] [Impact Index Per Article: 0] [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
|
3
|
Zhang Z, Jin L, Liu L, Zhou M, Zhang X, Zhang L. The intricate relationship between autoimmunity disease and neutrophils death patterns: a love-hate story. Apoptosis 2023; 28:1259-1284. [PMID: 37486407 DOI: 10.1007/s10495-023-01874-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2023] [Indexed: 07/25/2023]
Abstract
Autoimmune diseases are pathological conditions that result from the misidentification of self-antigens in immune system, leading to host tissue damage and destruction. These diseases can affect different organs and systems, including the blood, joints, skin, and muscles. Despite the significant progress made in comprehending the underlying pathogenesis, the complete mechanism of autoimmune disease is still not entirely understood. In autoimmune diseases, the innate immunocytes are not functioning properly: they are either abnormally activated or physically disabled. As a vital member of innate immunocyte, neutrophils and their modes of death are influenced by the microenvironment of different autoimmune diseases due to their short lifespan and diverse death modes. Related to neutrophil death pathways, apoptosis is the most frequent cell death form of neutrophil non-lytic morphology, delayed or aberrant apoptosis may contribute to the development anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis (AAV). In addition, NETosis, necroptosis and pyroptosis which are parts of lytic morphology exacerbate disease progression through various mechanisms in autoimmune diseases. This review aims to summarize recent advancements in understanding neutrophil death modes in various autoimmune diseases and provide insights into the development of novel therapeutic approaches for autoimmune diseases.
Collapse
Affiliation(s)
- Ziwei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China
| | - Lin Jin
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China
| | - Lianghu Liu
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China
| | - Mengqi Zhou
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China
| | - Xianzheng Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China.
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China.
| | - Lingling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, 230032, China.
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China.
- Anti-Inflammatory Immune Drugs Collaborative Innovation Center, Hefei, Anhui Province, China.
| |
Collapse
|
4
|
Nunoi H, Nakamura H, Nishimura T, Matsukura M. Recent topics and advanced therapies in chronic granulomatous disease. Hum Cell 2023; 36:515-527. [PMID: 36534309 DOI: 10.1007/s13577-022-00846-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022]
Abstract
Chronic granulomatous disease (CGD) is a primary immunodeficiency characterized by the inability of phagocytes to produce reactive oxygen species (ROS) owing to a defect in any of the five components (CYBB/gp91phox, CYBA/p22phox, NCF1/p47phox, NCF2/p67phox, and NCF4/p40phox) and a concomitant regulatory component of Rac1/2 and CYBC1/Eros of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase complex. Patients with CGD are at an increased risk of life-threatening infections caused by catalase-positive bacteria and fungi and of inflammatory complications such as CGD colitis. Antimicrobial and azole antifungal prophylaxes have considerably reduced the incidence and severity of bacterial and improved fungal infections and overall survival. CGD studies have revealed the precise epidemiology and role of NADPH oxidase in innate immunity which has led to a new understanding of the importance of phagocyte oxygen metabolism in various host-defense systems and the fields leading to cell death processes. Moreover, ROS plays central roles in the determination of cell fate as secondary messengers and by modifying of various signaling molecules. According to this increasing knowledge about the effects of ROS on the inflammasomal system, immunomodulatory treatments, such as IFN-γ and anti-IL-1 antibodies, have been established. This review covers the current topics in CGD and the relationship between ROS and ROS-mediated pathophysiological phenomena. In addition to the shirt summary of hematopoietic stem cell transplantation and gene therapy, we introduce a novel ROS-producing enzyme replacement therapy using PEG-fDAO to compensate for NADPH oxidase deficiency.
Collapse
Affiliation(s)
- Hiroyuki Nunoi
- Division of Pediatrics, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake-cho, Miyazaki-City, Miyazaki, 889-1692, Japan. .,Aisenkai Nichinan Hospital, 3649-2 Kazeta, Nichinan-City, Miyazaki, 887-0034, Japan.
| | - Hideki Nakamura
- Laboratory of Environmental Science and Technology, Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-Ku, Kumamoto City, 860-0082, Japan
| | - Toyoki Nishimura
- Division of Pediatrics, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake-cho, Miyazaki-City, Miyazaki, 889-1692, Japan
| | - Makoto Matsukura
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1 Ikeda, Nishi-Ku, Kumamoto City, 860-0082, Japan
| |
Collapse
|
5
|
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
|
6
|
Peng Z, Zhao C, Du X, Yang Y, Li Y, Song Y, Fang B, Zhang Y, Qin X, Zhang Y, Li X, Wang Z, Li X, Liu G. Autophagy Induced by Palmitic Acid Regulates Neutrophil Adhesion Through the Granule-Dependent Degradation of αMβ2 Integrin in Dairy Cows With Fatty Liver. Front Immunol 2021; 12:726829. [PMID: 34691032 PMCID: PMC8529007 DOI: 10.3389/fimmu.2021.726829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/02/2021] [Indexed: 11/28/2022] Open
Abstract
β2 integrins are critical for neutrophil firm adhesion, trans-endothelial migration, and the recruitment to the inflamed tissue. Autophagy is implicated in cell migration and tumor metastasis through facilitating the turnover of β1 integrins; however, whether autophagy is able to control neutrophil migration by promoting the degradation of β2 integrins is unexplored. Here, we show that high blood levels of palmitic acid (PA) strongly triggered neutrophil autophagy activation, leading to adhesion deficiency in dairy cows with fatty liver. The three neutrophil granule subtypes, namely, azurophil granules (AGs), specific granules (SGs), and gelatinase granules (GGs), were engulfed by the autophagosomes for degradation, resulting in an increased vacuolation in fatty liver dairy cow neutrophils. Importantly, the adhesion-associated molecules CD11b and CD18 distributed on AGs, SGs, and GGs were degraded with the three granule subtypes by autophagy. Moreover, FGA, Hsc70, and TRIM21 mediated the degradation of cytosolic oxidized–ubiquitinated CD11b and CD18. Collectively, our results demonstrate that high blood PA triggers neutrophil autophagy-dependent vacuolation and granule-dependent adhesion deficiency, decreasing neutrophil mobility, and impairing the innate immune system of dairy cow with fatty liver. This theory extends the category of autophagy in maintaining granule homeostasis and provides a novel strategy to improve the immune of dairy cows with metabolic disease.
Collapse
Affiliation(s)
- Zhicheng Peng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chenxu Zhao
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuchen Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yunfei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Baochen Fang
- College of Food Science and Engineering, Jilin University, Changchun, China
| | - Yuming Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xia Qin
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuanyuan Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiaobing Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
7
|
Theofani E, Xanthou G. Autophagy: A Friend or Foe in Allergic Asthma? Int J Mol Sci 2021; 22:ijms22126314. [PMID: 34204710 PMCID: PMC8231495 DOI: 10.3390/ijms22126314] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/04/2021] [Accepted: 06/10/2021] [Indexed: 12/20/2022] Open
Abstract
Autophagy is a major self-degradative process through which cytoplasmic material, including damaged organelles and proteins, are delivered and degraded in the lysosome. Autophagy represents a dynamic recycling system that produces new building blocks and energy, essential for cellular renovation, physiology, and homeostasis. Principal autophagy triggers include starvation, pathogens, and stress. Autophagy plays also a pivotal role in immune response regulation, including immune cell differentiation, antigen presentation and the generation of T effector responses, the development of protective immunity against pathogens, and the coordination of immunometabolic signals. A plethora of studies propose that both impaired and overactive autophagic processes contribute to the pathogenesis of human disorders, including infections, cancer, atherosclerosis, autoimmune and neurodegenerative diseases. Autophagy has been also implicated in the development and progression of allergen-driven airway inflammation and remodeling. Here, we provide an overview of recent studies pertinent to the biology of autophagy and molecular pathways controlling its activation, we discuss autophagy-mediated beneficial and detrimental effects in animal models of allergic diseases and illuminate new advances on the role of autophagy in the pathogenesis of human asthma. We conclude contemplating the potential of targeting autophagy as a novel therapeutic approach for the management of allergic responses and linked asthmatic disease.
Collapse
Affiliation(s)
- Efthymia Theofani
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 11547 Athens, Greece;
- 1st Department of Respiratory Medicine, “Sotiria” Regional Chest Diseases Hospital, Medical School, National Kapodistrian University of Athens, 11547 Athens, Greece
| | - Georgina Xanthou
- Cellular Immunology Laboratory, Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 11547 Athens, Greece;
- Correspondence: ; Tel.: +30-210-65-97-336
| |
Collapse
|
8
|
Increased TNF- α Initiates Cytoplasmic Vacuolization in Whole Blood Coculture with Dengue Virus. J Immunol Res 2021; 2021:6654617. [PMID: 34041302 PMCID: PMC8121593 DOI: 10.1155/2021/6654617] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 04/09/2021] [Accepted: 04/26/2021] [Indexed: 12/22/2022] Open
Abstract
During the acute febrile phase of dengue virus (DENV) infection, viremia can cause severe systemic immune responses accompanied by hematologic disorders. This study investigated the potential induction and mechanism of the cytopathic effects of DENV on peripheral blood cells ex vivo. At one day postinfection, there was viral nonstructural protein NS1 but no further virus replication measured in the whole blood culture. Notably, DENV exposure caused significant vacuolization in monocytic phagocytes. With a minor change in the complete blood cell count, except for a minor increase in neutrophils and a significant decrease in monocytes, the immune profiling assay identified several changes, particularly a significant reduction in CD14-positive monocytes as well as CD11c-positive dendritic cells. Abnormal production of TNF-α was highly associated with the induction of vacuolization. Manipulating TNF-α expression resulted in cytopathogenic effects. These results demonstrate the potential hematological damage caused by ex vivo DENV-induced TNF-α.
Collapse
|
9
|
Kono M, Matsuhiroya S, Obuchi A, Takahashi T, Imoto S, Kawano S, Saigo K. Deferasirox, an iron-chelating agent, alleviates acute lung inflammation by inhibiting neutrophil activation and extracellular trap formation. J Int Med Res 2021; 48:300060520951015. [PMID: 32938287 PMCID: PMC7503029 DOI: 10.1177/0300060520951015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE Reactive oxygen species (ROS) production by neutrophils induces pulmonary endothelial cell damage and results in acute lung injury (ALI). We previously reported that deferasirox (DFS), an iron-chelating agent, inhibits the ROS production and neutrophil extracellular trap (NET) formation induced by phorbol myristate acetate and formylmethionylleucylphenylalanine in vitro. In the present study, we investigated the effects of DFS in vivo using a mouse model of lipopolysaccharide (LPS)-induced ALI. METHODS After DFS administration for 7 days, ALI was induced in mice by LPS via intratracheal administration. RESULTS LPS treatment induced neutrophil invasion in the lung tissues, along with NET formation and a significant increase in the quantity of double-stranded DNA in the bronchoalveolar lavage fluid, while pre-administered DFS inhibited these phenomena. However, alteration of neutrophil morphology in the cytoplasm in terms of shape and vacuolization was not inhibited by the pre-administration of DFS, possibly through ROS production. CONCLUSIONS DFS suppressed neutrophil invasion into lung tissues and reduced the double-stranded DNA content released by the neutrophils. These results suggest that DFS can potentially be used to prevent diseases related to neutrophil activation including ALI, thrombosis, and vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Mari Kono
- Scientific Research, Scientific Affairs, Sysmex Corporation, Kobe, Japan
| | - Shiori Matsuhiroya
- Scientific Research, Scientific Affairs, Sysmex Corporation, Kobe, Japan
| | - Ayako Obuchi
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| | | | - Shion Imoto
- Department of Health Science, Kobe Tokiwa University, Kobe, Japan
| | - Seiji Kawano
- Integrated Clinical Education Center, Kobe University Hospital, Kobe, Japan
| | - Katsuyasu Saigo
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| |
Collapse
|
10
|
Al-Aamri HM, Irving HR, Bradley C, Meehan-Andrews T. Intrinsic and extrinsic apoptosis responses in leukaemia cells following daunorubicin treatment. BMC Cancer 2021; 21:438. [PMID: 33879127 PMCID: PMC8059319 DOI: 10.1186/s12885-021-08167-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 04/06/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Daunorubicin is used clinically in the treatment of myeloma, acute lymphatic and myelocytic leukaemia. The toxic lesions caused by daunorubicin induce various modes of cell death, including apoptosis. Apoptosis is highly regulated programmed cell death that can be initiated mainly via two pathways, through death receptors (extrinsic) or involvement of the mitochondria (intrinsic). Induction of apoptosis via these pathways has been alluded following treatment with daunorubicin, but never compared in acute lymphoblastic leukaemia over a time course. METHODS This study investigated the mechanisms of daunorubicin induced apoptosis in the treatment of CCRF-CEM, MOLT-4 (acute T-lymphoblastic leukaemia) and SUP-B15 (acute B-lymphoblastic leukaemia) cells. Cells were treated with daunorubicin for 4 h, and then placed in recovery medium (without daunorubicin) for 4 h, 12 h and 24 h. Apoptotic response was analysing using annexin-V expression, caspase activity, mitochondrial membrane potential change and an array to detect 43 apoptotic proteins. RESULTS Daunorubicin induced apoptosis in all leukemic cell lines, but with different levels and duration of response. Both apoptosis levels and caspase activity increased after four hours recovery then declined in CCRF-CEM and MOLT-4 cells. However, SUP-B15 cells displayed initially comparable levels but remained elevated over the 24 h assessment period. Changes in mitochondrial membrane potential occurred in both MOLT-4 and CCRF-CEM cells but not in SUP-B15 cells. Expression of apoptotic proteins, including Bcl-2, Bax, caspase 3 and FADD, indicated that daunorubicin potentially induced both extrinsic and intrinsic apoptosis in both CCRF-CEM and MOLT-4 cells, but only extrinsic apoptosis in SUP-B15 cells. CONCLUSIONS This study describes variations in sensitivities and timing of apoptotic responses in different leukaemia cell lines. These differences could be attributed to the lack of functional p53 in coordinating the cells response following cytotoxic treatment with daunorubicin, which appears to delay apoptosis and utilises alternative signalling mechanisms that need to be further explored.
Collapse
Affiliation(s)
- Hussain Mubarak Al-Aamri
- Department of Pharmacy and Biomedical Science, La Trobe Institute for Molecular Science (LIMS), La Trobe University, P.O. Box 199, Bendigo, VIC, 3552, Australia.,Oman College of Health Sciences, PO Box 293, 620, Ruwi, Sultanate of Oman
| | - Helen R Irving
- Department of Pharmacy and Biomedical Science, La Trobe Institute for Molecular Science (LIMS), La Trobe University, P.O. Box 199, Bendigo, VIC, 3552, Australia
| | - Christopher Bradley
- Department of Pharmacy and Biomedical Science, La Trobe Institute for Molecular Science (LIMS), La Trobe University, P.O. Box 199, Bendigo, VIC, 3552, Australia
| | - Terri Meehan-Andrews
- Department of Pharmacy and Biomedical Science, La Trobe Institute for Molecular Science (LIMS), La Trobe University, P.O. Box 199, Bendigo, VIC, 3552, Australia.
| |
Collapse
|
11
|
Allaeys I, Ribeiro de Vargas F, Bourgoin SG, Poubelle PE. Human Inflammatory Neutrophils Express Genes Encoding Peptidase Inhibitors: Production of Elafin Mediated by NF-κB and CCAAT/Enhancer-Binding Protein β. THE JOURNAL OF IMMUNOLOGY 2021; 206:1943-1956. [PMID: 33762327 DOI: 10.4049/jimmunol.2000852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 02/11/2021] [Indexed: 11/19/2022]
Abstract
The concept of plasticity of neutrophils is highlighted by studies showing their ability to transdifferentiate into APCs. In this regard, transdifferentiated neutrophils were found at inflammatory sites of autoimmune arthritis (AIA). Exposure of neutrophils to inflammatory stimuli prolongs their survival, thereby favoring the acquisition of pathophysiologically relevant phenotypes and functions. By using microarrays, quantitative RT-PCR, and ELISAs, we showed that long-lived (LL) neutrophils obtained after 48 h of culture in the presence of GM-CSF, TNF, and IL-4 differentially expressed genes related to apoptosis, MHC class II, immune response, and inflammation. The expression of anti-inflammatory genes mainly of peptidase inhibitor families is upregulated in LL neutrophils. Among these, the PI3 gene encoding elafin was the most highly expressed. The de novo production of elafin by LL neutrophils depended on a synergism between GM-CSF and TNF via the activation and cooperativity of C/EBPβ and NF-κB pathways, respectively. Elafin concentrations were higher in synovial fluids (SF) of patients with AIA than in SF of osteoarthritis. SF neutrophils produced more elafin than blood counterparts. These results are discussed with respect to implications of neutrophils in chronic inflammation and the potential influence of elafin in AIA.
Collapse
Affiliation(s)
- Isabelle Allaeys
- Infectious Diseases and Immunity Research Division, Department of Medicine, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, Quebec G1V 4G2, Canada
| | - Flavia Ribeiro de Vargas
- Infectious Diseases and Immunity Research Division, Department of Medicine, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, Quebec G1V 4G2, Canada
| | - Sylvain G Bourgoin
- Infectious Diseases and Immunity Research Division, Department of Medicine, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, Quebec G1V 4G2, Canada
| | - Patrice E Poubelle
- Infectious Diseases and Immunity Research Division, Department of Medicine, Centre Hospitalier Universitaire de Québec-Université Laval Research Center, Quebec City, Quebec G1V 4G2, Canada
| |
Collapse
|
12
|
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: 91] [Impact Index Per Article: 22.8] [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
|
13
|
Wang X, Avsec D, Obreza A, Yousefi S, Mlinarič-Raščan I, Simon HU. A Putative Serine Protease is Required to Initiate the RIPK3-MLKL-Mediated Necroptotic Death Pathway in Neutrophils. Front Pharmacol 2021; 11:614928. [PMID: 33551816 PMCID: PMC7860068 DOI: 10.3389/fphar.2020.614928] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
Adhesion receptors, such as CD44, have been shown to activate receptor interacting protein kinase-3 (RIPK3)—mixed lineage kinase-like (MLKL) signaling, leading to a non-apoptotic cell death in human granulocyte/macrophage colony-stimulating factor (GM-CSF) – primed neutrophils. The signaling events of this necroptotic pathway, however, remain to be investigated. In the present study, we report the design, synthesis, and characterization of a series of novel serine protease inhibitors. Two of these inhibitors, compounds 1 and 3, were able to block CD44-triggered necroptosis in GM-CSF-primed neutrophils. Both inhibitors prevented the activation of MLKL, p38 mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3’—kinase (PI3K), hence blocking the increased levels of reactive oxygen species (ROS) required for cell death. Although compounds one and three partially inhibited isolated human neutrophil elastase (HNE) activity, we obtained no pharmacological evidence that HNE is involved in the initiation of this death pathway within a cellular context. Interestingly, neither serine protease inhibitor had any effect on FAS receptor-mediated apoptosis. Taken together, these results suggest that a serine protease is involved in non-apoptotic CD44-triggered RIPK3-MLKL-dependent neutrophil cell death, but not FAS receptor-mediated caspase-dependent apoptosis. Thus, a pharmacological block on serine proteases might be beneficial for preventing exacerbation of disease in neutrophilic inflammatory responses.
Collapse
Affiliation(s)
- Xiaoliang Wang
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, Bern, Switzerland
| | - Damjan Avsec
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Aleš Obreza
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, Bern, Switzerland
| | | | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, Bern, Switzerland.,Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| |
Collapse
|
14
|
Wehrli M, Schneider C, Cortinas-Elizondo F, Verschoor D, Frias Boligan K, Adams OJ, Hlushchuk R, Engelmann C, Daudel F, Villiger PM, Seibold F, Yawalkar N, Vonarburg C, Miescher S, Lötscher M, Kaufmann T, Münz C, Mueller C, Djonov V, Simon HU, von Gunten S. IgA Triggers Cell Death of Neutrophils When Primed by Inflammatory Mediators. THE JOURNAL OF IMMUNOLOGY 2020; 205:2640-2648. [PMID: 33008951 DOI: 10.4049/jimmunol.1900883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
IVIG preparations consisting of pooled IgG are increasingly used for the treatment of autoimmune diseases. IVIG is known to regulate the viability of immune cells, including neutrophils. We report that plasma-derived IgA efficiently triggers death of neutrophils primed by cytokines or TLR agonists. IgA-mediated programmed neutrophil death was PI3K-, p38 MAPK-, and JNK-dependent and evoked anti-inflammatory cytokines in macrophage cocultures. Neutrophils from patients with acute Crohn's disease, rheumatoid arthritis, or sepsis were susceptible to both IgA- and IVIG-mediated death. In contrast to IVIG, IgA did not promote cell death of quiescent neutrophils. Our findings suggest that plasma-derived IgA might provide a therapeutic option for the treatment of neutrophil-associated inflammatory disorders.
Collapse
Affiliation(s)
- Marc Wehrli
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | | | | | | | | | - Olivia Joan Adams
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Ruslan Hlushchuk
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Christine Engelmann
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Fritz Daudel
- Intensive Care Unit, Spital Thun, 3600 Thun, Switzerland
| | - Peter M Villiger
- Department of Rheumatology/Clinical Immunology/Allergology, University Hospital Bern, 3008 Bern, Switzerland
| | - Frank Seibold
- Gastroenterologie, Spitalnetz Bern, 3004 Bern, Switzerland.,Gastroenterologie, Praxis Balsiger, Seibold und Partner am Lindenhofspital, 3012 Bern, Switzerland
| | - Nikhil Yawalkar
- Department of Dermatology, University Hospital Bern, University of Bern, 3010 Bern, Switzerland
| | | | | | | | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Christoph Mueller
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; and
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland.,Department of Clinical Immunology and Allergology, Sechenov University, Moscow 119991, Russia
| | | |
Collapse
|
15
|
Wang X, Gessier F, Perozzo R, Stojkov D, Hosseini A, Amirshahrokhi K, Kuchen S, Yousefi S, Lötscher P, Simon HU. RIPK3–MLKL–Mediated Neutrophil Death Requires Concurrent Activation of Fibroblast Activation Protein-α. THE JOURNAL OF IMMUNOLOGY 2020; 205:1653-1663. [DOI: 10.4049/jimmunol.2000113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 07/21/2020] [Indexed: 12/30/2022]
|
16
|
Painter JD, Galle-Treger L, Akbari O. Role of Autophagy in Lung Inflammation. Front Immunol 2020; 11:1337. [PMID: 32733448 PMCID: PMC7358431 DOI: 10.3389/fimmu.2020.01337] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
Autophagy is a cellular recycling system found in almost all types of eukaryotic organisms. The system is made up of a variety of proteins which function to deliver intracellular cargo to lysosomes for formation of autophagosomes in which the contents are degraded. The maintenance of cellular homeostasis is key in the survival and function of a variety of human cell populations. The interconnection between metabolism and autophagy is extensive, therefore it has a role in a variety of different cell functions. The disruption or dysfunction of autophagy in these cell types have been implicated in the development of a variety of inflammatory diseases including asthma. The role of autophagy in non-immune and immune cells both lead to the pathogenesis of lung inflammation. Autophagy in pulmonary non-immune cells leads to tissue remodeling which can develop into chronic asthma cases with long term effects. The role autophagy in the lymphoid and myeloid lineages in the pathology of asthma differ in their functions. Impaired autophagy in lymphoid populations have been shown, in general, to decrease inflammation in both asthma and inflammatory disease models. Many lymphoid cells rely on autophagy for effector function and maintained inflammation. In stark contrast, autophagy deficient antigen presenting cells have been shown to have an activated inflammasome. This is largely characterized by a TH17 response that is accompanied with a much worse prognosis including granulocyte mediated inflammation and steroid resistance. The cell specificity associated with changes in autophagic flux complicates its targeting for amelioration of asthmatic symptoms. Differing asthmatic phenotypes between TH2 and TH17 mediated disease may require different autophagic modulations. Therefore, treatments call for a more cell specific and personalized approach when looking at chronic asthma cases. Viral-induced lung inflammation, such as that caused by SARS-CoV-2, also may involve autophagic modulation leading to inflammation mediated by lung resident cells. In this review, we will be discussing the role of autophagy in non-immune cells, myeloid cells, and lymphoid cells for their implications into lung inflammation and asthma. Finally, we will discuss autophagy's role viral pathogenesis, immunometabolism, and asthma with insights into autophagic modulators for amelioration of lung inflammation.
Collapse
Affiliation(s)
- Jacob D Painter
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Lauriane Galle-Treger
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
17
|
Shrestha S, Lee JM, Hong CW. Autophagy in neutrophils. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2020; 24:1-10. [PMID: 31908569 PMCID: PMC6940497 DOI: 10.4196/kjpp.2020.24.1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 02/06/2023]
Abstract
Autophagy is a highly conserved intracellular degradation and energy-recycling mechanism that contributes to the maintenance of cellular homeostasis. Extensive researches over the past decades have defined the role of autophagy innate immune cells. In this review, we describe the current state of knowledge regarding the role of autophagy in neutrophil biology and a picture of molecular mechanism underlying autophagy in neutrophils. Neutrophils are professional phagocytes that comprise the first line of defense against pathogen. Autophagy machineries are highly conserved in neutrophils. Autophagy is not only involved in generalized function of neutrophils such as differentiation in bone marrow but also plays crucial role effector functions of neutrophils such as granule formation, degranulation, neutrophil extracellular traps release, cytokine production, bactericidal activity and controlling inflammation. This review outlines the current understanding of autophagy in neutrophils and provides insight towards identification of novel therapeutics targeting autophagy in neutrophils.
Collapse
Affiliation(s)
- Sanjeeb Shrestha
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Jae Man Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Chang-Won Hong
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| |
Collapse
|
18
|
Fert-Bober J, Darrah E, Andrade F. Insights into the study and origin of the citrullinome in rheumatoid arthritis. Immunol Rev 2019; 294:133-147. [PMID: 31876028 DOI: 10.1111/imr.12834] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/08/2019] [Indexed: 12/11/2022]
Abstract
The presence of autoantibodies and autoreactive T cells to citrullinated proteins and citrullinating enzymes in patients with rheumatoid arthritis (RA), together with the accumulation of citrullinated proteins in rheumatoid joints, provides substantial evidence that dysregulated citrullination is a hallmark feature of RA. However, understanding mechanisms that dysregulate citrullination in RA has important challenges. Citrullination is a normal process in immune and non-immune cells, which is likely activated by different conditions (eg, inflammation) with no pathogenic consequences. In a complex inflammatory environment such as the RA joint, unique strategies are therefore required to dissect specific mechanisms involved in the abnormal production of citrullinated proteins. Here, we will review current models of citrullination in RA and discuss critical components that, in our view, are relevant to understanding the accumulation of citrullinated proteins in the RA joint, collectively referred to as the RA citrullinome. In particular, we will focus on potential caveats in the study of citrullination in RA and will highlight methods to precisely detect citrullinated proteins in complex biological samples, which is a confirmatory approach to mechanistically link the RA citrullinome with unique pathogenic pathways in RA.
Collapse
Affiliation(s)
- Justyna Fert-Bober
- The Smidt Heart Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Erika Darrah
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Felipe Andrade
- Division of Rheumatology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Gößwein S, Lindemann A, Mahajan A, Maueröder C, Martini E, Patankar J, Schett G, Becker C, Wirtz S, Naumann-Bartsch N, Bianchi ME, Greer PA, Lochnit G, Herrmann M, Neurath MF, Leppkes M. Citrullination Licenses Calpain to Decondense Nuclei in Neutrophil Extracellular Trap Formation. Front Immunol 2019; 10:2481. [PMID: 31695698 PMCID: PMC6817590 DOI: 10.3389/fimmu.2019.02481] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 10/04/2019] [Indexed: 12/23/2022] Open
Abstract
Neutrophils respond to various stimuli by decondensing and releasing nuclear chromatin characterized by citrullinated histones as neutrophil extracellular traps (NETs). This achieves pathogen immobilization or initiation of thrombosis, yet the molecular mechanisms of NET formation remain elusive. Peptidyl arginine deiminase-4 (PAD4) achieves protein citrullination and has been intricately linked to NET formation. Here we show that citrullination represents a major regulator of proteolysis in the course of NET formation. Elevated cytosolic calcium levels trigger both peptidylarginine deiminase-4 (PAD4) and calpain activity in neutrophils resulting in nuclear decondensation typical of NETs. Interestingly, PAD4 relies on proteolysis by calpain to achieve efficient nuclear lamina breakdown and chromatin decondensation. Pharmacological or genetic inhibition of PAD4 and calpain strongly inhibit chromatin decondensation of human and murine neutrophils in response to calcium ionophores as well as the proteolysis of nuclear proteins like lamin B1 and high mobility group box protein 1 (HMGB1). Taken together, the concerted action of PAD4 and calpain induces nuclear decondensation in the course of calcium-mediated NET formation.
Collapse
Affiliation(s)
- Stefanie Gößwein
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, Kussmaul Campus for Medical Research and Translational Research Center, Erlangen, Germany
| | - Aylin Lindemann
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, Kussmaul Campus for Medical Research and Translational Research Center, Erlangen, Germany
| | - Aparna Mahajan
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christian Maueröder
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Eva Martini
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, Kussmaul Campus for Medical Research and Translational Research Center, Erlangen, Germany
| | - Jay Patankar
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, Kussmaul Campus for Medical Research and Translational Research Center, Erlangen, Germany
| | - Georg Schett
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, Kussmaul Campus for Medical Research and Translational Research Center, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, Kussmaul Campus for Medical Research and Translational Research Center, Erlangen, Germany
| | - Nora Naumann-Bartsch
- Department of Pediatrics, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Marco E Bianchi
- Chromatin Dynamics Unit, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Peter A Greer
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Günter Lochnit
- Institute of Biochemistry, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Martin Herrmann
- Department of Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, Kussmaul Campus for Medical Research and Translational Research Center, Erlangen, Germany
| | - Moritz Leppkes
- Department of Medicine 1, Friedrich-Alexander-Universität Erlangen-Nürnberg, Deutsches Zentrum Immuntherapie, Kussmaul Campus for Medical Research and Translational Research Center, Erlangen, Germany
| |
Collapse
|
20
|
Talla U, Bozonet SM, Parker HA, Hampton MB, Vissers MCM. Prolonged exposure to hypoxia induces an autophagy-like cell survival program in human neutrophils. J Leukoc Biol 2019; 106:1367-1379. [PMID: 31412152 DOI: 10.1002/jlb.4a0319-079rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/14/2019] [Accepted: 07/14/2019] [Indexed: 12/13/2022] Open
Abstract
Neutrophils contribute to low oxygen availability at inflammatory sites through the generation of reactive oxidants. They are also functionally affected by hypoxia, which delays neutrophil apoptosis. However, the eventual fate of neutrophils in hypoxic conditions is unknown and this is important for their effective clearance and the resolution of inflammation. We have monitored the survival and function of normal human neutrophils exposed to hypoxia over a 48 h period. Apoptosis was delayed, and the cells remained intact even at 48 h. However, hypoxia promoted significant changes in neutrophil morphology with the appearance of many new cytoplasmic vesicles, often containing cell material, within 5 hours of exposure to low O2 . This coincided with an increase in LC3B-II expression, indicative of autophagosome formation and an autophagy-like process. In hypoxic conditions, neutrophils preferentially lost myeloperoxidase, a marker of azurophil granules. Short-term (2 h) hypoxic exposure resulted in sustained potential to generate superoxide when O2 was restored, but the capacity for oxidant production was lost with longer periods of hypoxia. Phagocytic ability was unchanged by hypoxia, and bacterial killing by neutrophils in both normoxic and hypoxic conditions was substantially diminished after 24 hours. However, pre-exposure to hypoxia resulted in an enhanced ability to kill bacteria by oxidant-independent mechanisms. Our data provide the first evidence for hypoxia as a driver of neutrophil autophagy that can influence the function and ultimate fate of these cells, including their eventual clearance and the resolution of inflammation.
Collapse
Affiliation(s)
- Usharani Talla
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Stephanie M Bozonet
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Heather A Parker
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Mark B Hampton
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Margreet C M Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| |
Collapse
|
21
|
Frangou E, Vassilopoulos D, Boletis J, Boumpas DT. An emerging role of neutrophils and NETosis in chronic inflammation and fibrosis in systemic lupus erythematosus (SLE) and ANCA-associated vasculitides (AAV): Implications for the pathogenesis and treatment. Autoimmun Rev 2019; 18:751-760. [DOI: 10.1016/j.autrev.2019.06.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/02/2019] [Indexed: 02/08/2023]
|
22
|
Abstract
In addition to their well-known role as the cellular mediators of immunity, key other roles have been identified for neutrophils during septic shock. Importantly, neutrophils indeed play a critical role in the recently described immunothrombosis concept and in septic shock-induced coagulopathy. Septic shock is one of the most severe forms of infection, characterized by an inadequate host response to the pathogenic organism. This host response involves numerous defense mechanisms with an intense cellular activation, including neutrophil activation. Neutrophils are key cells of innate immunity through complex interactions with vascular cells and their activation may participate in systemic tissue damages. Their activation also leads to the emission of neutrophil extracellular traps, which take part in both pathogen circumscription and phagocytosis, but also in coagulation activation. Neutrophils thus stand at the interface between hemostasis and immunity, called immunothrombosis.The present review will develop a cellular approach of septic shock pathophysiology focusing on neutrophils as key players of septic shock-induced vascular cell dysfunction and of the host response, associating immunity and hemostasis. We will therefore first develop the role of neutrophils in the interplay between innate and adaptive immunity, and will then highlight recent advances in our understanding of immunothrombosis septic shock-induced coagulopathy.
Collapse
|
23
|
Boeltz S, Amini P, Anders HJ, Andrade F, Bilyy R, Chatfield S, Cichon I, Clancy DM, Desai J, Dumych T, Dwivedi N, Gordon RA, Hahn J, Hidalgo A, Hoffmann MH, Kaplan MJ, Knight JS, Kolaczkowska E, Kubes P, Leppkes M, Manfredi AA, Martin SJ, Maueröder C, Maugeri N, Mitroulis I, Munoz LE, Nakazawa D, Neeli I, Nizet V, Pieterse E, Radic MZ, Reinwald C, Ritis K, Rovere-Querini P, Santocki M, Schauer C, Schett G, Shlomchik MJ, Simon HU, Skendros P, Stojkov D, Vandenabeele P, Berghe TV, van der Vlag J, Vitkov L, von Köckritz-Blickwede M, Yousefi S, Zarbock A, Herrmann M. To NET or not to NET:current opinions and state of the science regarding the formation of neutrophil extracellular traps. Cell Death Differ 2019; 26:395-408. [PMID: 30622307 PMCID: PMC6370810 DOI: 10.1038/s41418-018-0261-x] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 11/05/2018] [Accepted: 11/30/2018] [Indexed: 12/16/2022] Open
Abstract
Since the discovery and definition of neutrophil extracellular traps (NETs) 14 years ago, numerous characteristics and physiological functions of NETs have been uncovered. Nowadays, the field continues to expand and novel mechanisms that orchestrate formation of NETs, their previously unknown properties, and novel implications in disease continue to emerge. The abundance of available data has also led to some confusion in the NET research community due to contradictory results and divergent scientific concepts, such as pro- and anti-inflammatory roles in pathologic conditions, demarcation from other forms of cell death, or the origin of the DNA that forms the NET scaffold. Here, we present prevailing concepts and state of the science in NET-related research and elaborate on open questions and areas of dispute.
Collapse
Affiliation(s)
- Sebastian Boeltz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Poorya Amini
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Felipe Andrade
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rostyslav Bilyy
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Simon Chatfield
- Inflammation Division, Walter and Eliza Hall Institute, Melbourne, Victoria, Australia
| | - Iwona Cichon
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Danielle M Clancy
- VIB-UGent Center for Inflammation Research, University of Gent, Gent, Belgium
| | - Jyaysi Desai
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Tetiana Dumych
- Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Nishant Dwivedi
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rachael Ann Gordon
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jonas Hahn
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Andrés Hidalgo
- Department of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares (CNIC) Carlos III, Madrid, Spain
- Institute for Cardiovascular Prevention, Ludwig Maximilians University, Munich, Germany
| | - Markus H Hoffmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany.
| | - Mariana J Kaplan
- Systemic Autoimmunity Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, USA
| | - Jason S Knight
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Elzbieta Kolaczkowska
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Paul Kubes
- Snyder institute of Chronic Diseases, University of Calgary, Calgary, Canada
| | - Moritz Leppkes
- Department of Medicine 1 - Gastroenterology, Pulmonology and Endocrinology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Angelo A Manfredi
- Università Vita Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy
| | - Seamus J Martin
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Christian Maueröder
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
- VIB-UGent Center for Inflammation Research, University of Gent, Gent, Belgium
| | - Norma Maugeri
- Università Vita Salute San Raffaele and IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ioannis Mitroulis
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Luis E Munoz
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Daigo Nakazawa
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Indira Neeli
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Victor Nizet
- UC San Diego School of Medicine, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UC San Diego, La Jolla, CA, USA
| | - Elmar Pieterse
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marko Z Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Christiane Reinwald
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Konstantinos Ritis
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Michal Santocki
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Christine Schauer
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| | - Mark Jay Shlomchik
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia
| | - Panagiotis Skendros
- Laboratory of Molecular Hematology, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, University of Gent, Gent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Methusalem platform, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, University of Gent, Gent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Pathophysiology, Faculty of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ljubomir Vitkov
- Department of Biosciences, Vascular & Exercise Biology Unit, University of Salzburg, Salzburg, Austria
- Periodontology and Preventive Dentistry, Saarland University, Homburg, Germany
| | - Maren von Köckritz-Blickwede
- Department of Physiological Chemistry & Research Center for Emerging Infections and Zoonosis (RIZ), University of Veterinary Medicine Hannover, Hannover, Germany
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Alexander Zarbock
- University of Münster, Department of Anesthesiology, Intensive Care and Pain Medicine, Münster, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University (FAU) Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054, Erlangen, Germany
| |
Collapse
|
24
|
Germic N, Frangez Z, Yousefi S, Simon HU. Regulation of the innate immune system by autophagy: neutrophils, eosinophils, mast cells, NK cells. Cell Death Differ 2019; 26:703-714. [PMID: 30737478 PMCID: PMC6460399 DOI: 10.1038/s41418-019-0295-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 12/15/2022] Open
Abstract
Autophagy is an evolutionally conserved, highly regulated catabolic process that combines cellular functions required for the regulation of metabolic balance under conditions of stress with those needed for the degradation of damaged cell organelles via the lysosomal machinery. The importance of autophagy for cell homeostasis and survival has long been appreciated. Recent data suggest that autophagy is also involved in non-metabolic functions that impact the immune system. Here, we reflect in two review articles the recent literature pointing to an important role for autophagy in innate immune cells. In this article, we focus on neutrophils, eosinophils, mast cells, and natural killer cells. We mainly discuss the influence of autophagy on functional cellular responses and its importance for overall host defense. In the companion review, we present the role of autophagy in the functions performed by monocytes/macrophages and dendritic cells.
Collapse
Affiliation(s)
- Nina Germic
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Ziva Frangez
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
- Department of Clinical Immunology and Allergology, Sechenov University, Moscow, Russia.
| |
Collapse
|
25
|
Ramesh J, Ronsard L, Gao A, Venugopal B. Autophagy Intertwines with Different Diseases-Recent Strategies for Therapeutic Approaches. Diseases 2019; 7:diseases7010015. [PMID: 30717078 PMCID: PMC6473623 DOI: 10.3390/diseases7010015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 01/22/2019] [Accepted: 01/28/2019] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a regular and substantial “clear-out process” that occurs within the cell and that gets rid of debris that accumulates in membrane-enclosed vacuoles by using enzyme-rich lysosomes, which are filled with acids that degrade the contents of the vacuoles. This machinery is well-connected with many prevalent diseases, including cancer, HIV, and Parkinson’s disease. Considering that autophagy is well-known for its significant connections with a number of well-known fatal diseases, a thorough knowledge of the current findings in the field is essential in developing therapies to control the progression rate of diseases. Thus, this review summarizes the critical events comprising autophagy in the cellular system and the significance of its key molecules in manifesting this pathway in various diseases for down- or upregulation. We collectively reviewed the role of autophagy in various diseases, mainly neurodegenerative diseases, cancer, inflammatory diseases, and renal disorders. Here, some collective reports on autophagy showed that this process might serve as a dual performer: either protector or contributor to certain diseases. The aim of this review is to help researchers to understand the role of autophagy-regulating genes encoding functional open reading frames (ORFs) and its connection with diseases, which will eventually drive better understanding of both the progression and suppression of different diseases at various stages. This review also focuses on certain novel therapeutic strategies which have been published in the recent years based on targeting autophagy key proteins and its interconnecting signaling cascades.
Collapse
Affiliation(s)
- Janani Ramesh
- Department of Medical Biochemistry, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India.
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Larance Ronsard
- The Ragon Institute of Massachusetts General Hospital, The Massachusetts Institute of Technology and Harvard University, 400 Technology Square, Cambridge, MA 02140, USA.
| | - Anthony Gao
- Renal Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bhuvarahamurthy Venugopal
- Department of Medical Biochemistry, Dr. A.L.M. Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India.
| |
Collapse
|
26
|
Dąbrowska D, Jabłońska E, Iwaniuk A, Garley M. Many Ways-One Destination: Different Types of Neutrophils Death. Int Rev Immunol 2018; 38:18-32. [PMID: 30516403 DOI: 10.1080/08830185.2018.1540616] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Neutrophils constitute the most numerous populations of peripheral blood leukocytes, fulfilling the fundamental role in the development of the innate immune response. As the cells of the first line of defense, they guard the organism against the spread of pathogenic microorganisms. Neutrophils, similar to the other cells of the immune system, enter the path of death after fulfilling their biological function. Depending on the conditions that they are found in, they may undergo different types of cell death which requires the involvement of numerous signaling pathways. In this review article, we summarize the current state of knowledge regarding the different forms of neutrophil death, such as apoptosis, necrosis, necroptosis, autophagy, NETosis and pyroptosis.
Collapse
Affiliation(s)
- Dorota Dąbrowska
- a Department of Immunology , Medical University of Bialystok , Bialystok , Poland
| | - Ewa Jabłońska
- a Department of Immunology , Medical University of Bialystok , Bialystok , Poland
| | - Agnieszka Iwaniuk
- a Department of Immunology , Medical University of Bialystok , Bialystok , Poland
| | - Marzena Garley
- a Department of Immunology , Medical University of Bialystok , Bialystok , Poland
| |
Collapse
|
27
|
Kono M, Saigo K, Matsuhiroya S, Takahashi T, Hashimoto M, Obuchi A, Imoto S, Nishiyama T, Kawano S. Detection of activated neutrophils by reactive oxygen species production using a hematology analyzer. J Immunol Methods 2018; 463:122-126. [PMID: 30339797 DOI: 10.1016/j.jim.2018.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 10/05/2018] [Accepted: 10/11/2018] [Indexed: 10/28/2022]
Abstract
Neutrophils are recruited to infection sites and kill bacteria by phagocytosis and reactive oxygen species (ROS) production. It has been reported that vacuoles are present in neutrophils that produce ROS and are present in large numbers in blood smears of patients with bacterial infections. The leukocyte differentiation function on the Sysmex automated hematology analyzer classifies leukocytes by flow cytometry. Particularly, side-scattered light is known to reflect the quantity of organelles. This study investigated the possibility of detecting vacuoles or invagination of cell membrane in neutrophils producing ROS using a hematology analyzer. Whole blood and polymorphonuclear (PMN) cell fractions were activated with phorbol myristate acetate (PMA) or formylmethionylleucylphenylalanine (fMLP) and analyzed using the Sysmex XE-2100 automated hematology analyzer. PMN fractions were morphologically analyzed with a confocal laser scanning microscope (CLSM), electron microscope (EM), and general-purpose conventional flow cytometer. In the white blood cell differentiation scattergram obtained in this analysis, a new cluster separate from the original neutrophil cluster appeared in the eosinophil area in an area of higher side-scattering (SSC) intensity. Flow cytometry analysis of the PMN fractions revealed that the cells in this new cluster were CD16b- and APF-positive, indicating that the cells were activated neutrophils that produced ROS. CLSM and EM findings revealed that ROS production occurred in the cytoplasm and that the activated neutrophils contained some vacuole-like structures of vacuoles or invagination of cell membrane. Vacuole-like Sstructures were found within the cytoplasm of neutrophils producing ROS. These neutrophils were detected as an independent cluster in the eosinophil area with higher SSC intensity than that shown by neutrophils in the traditional cluster on the white blood cell differentiation scattergram, likely because the vacuole-like structures increased the SSC intensity.
Collapse
Affiliation(s)
- Mari Kono
- Scientific Research, Scientific Affairs, Sysmex Corporation, Kobe, Japan.
| | - Katsuyasu Saigo
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| | - Shiori Matsuhiroya
- Scientific Research, Scientific Affairs, Sysmex Corporation, Kobe, Japan
| | | | | | - Ayako Obuchi
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| | - Shion Imoto
- Department of Health Science, Kobe Tokiwa University, Kobe, Japan
| | | | | |
Collapse
|
28
|
Barbosa LA, Fiuza PP, Borges LJ, Rolim FA, Andrade MB, Luz NF, Quintela-Carvalho G, Lima JB, Almeida RP, Chan FK, Bozza MT, Borges VM, Prates DB. RIPK1-RIPK3-MLKL-Associated Necroptosis Drives Leishmania infantum Killing in Neutrophils. Front Immunol 2018; 9:1818. [PMID: 30154785 PMCID: PMC6102393 DOI: 10.3389/fimmu.2018.01818] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022] Open
Abstract
Necroptosis is a pro-inflammatory cell death, which happens in the context of caspase-8 inhibition, allowing activation of the receptor interacting protein kinase 1-receptor interacting protein kinase 3-mixed lineage kinase domain-like (RIPK1-RIPK3-MLKL) axis. Recently, necroptosis has emerged as a key component of resistance against pathogens including infected macrophage by Leishmania infantum, the ethiologic agent of Visceral leishmaniasis (VL). VL is the most severe form of Leishmaniasis, characterized by systemic inflammation and neutropenia. However, the role of neutrophil cell death in VL has not been characterized. Here, we showed that VL patients exhibited increased lactate dehydrogenase levels in the serum, a hallmark of cell death and tissue damage. We investigated the effect of necroptosis in neutrophil infection in vitro. Human neutrophils pretreated with zVAD-fmk (pan-caspase inhibitor) and zIETD-fmk (caspase-8 inhibitor) increased reactive oxygen species (ROS) level in response to Leishmania infection, which is associated with necroptotic cell death. MLKL, an important effector molecule downstream of necroptosis pathway, was also required for Leishmania killing. Moreover, in absence of caspases-8, murine neutrophils displayed loss of membrane integrity, higher levels of ROS, and decreased L. infantum viability. Pharmacological inhibition of RIPK1 or RIPK3 increased parasite survival when caspase-8 was blocked. Electron microscopy assays revealed morphological features associated with necroptotic death in L. infantum infected-neutrophils pretreated with caspase inhibitor, whereas infected cells pretreated with RIPK1 and RIPK3 inhibitors did not show ultra-structural alterations in membrane integrity and presented viable Leishmania within parasitophorous vacuoles. Taken together, these findings suggest that inhibition of caspase-8 contributes to elimination of L. infantum in neutrophils by triggering necroptosis. Thus, targeting necroptosis may represent a new strategy to control Leishmania replication.
Collapse
Affiliation(s)
| | | | | | | | | | - Nivea F Luz
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Graziele Quintela-Carvalho
- Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Instituto Federal de Educação, Ciência e Tecnologia Baiano, Santa Inês, Brazil
| | - Jonilson B Lima
- Centro de Ciências Biológicas e da Saúde, Universidade do Oeste da Bahia, Barreiras, Brazil
| | - Roque P Almeida
- Departamento de Medicina, Universidade Federal de Sergipe, Aracaju, Brazil
| | - Francis K Chan
- Department of Pathology, Immunology and Microbiology Program, University of Massachusetts Medical School, Worcester, MA, United States
| | - Marcelo T Bozza
- Departamento de Imunologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Valeria M Borges
- Universidade Federal da Bahia, Salvador, Brazil.,Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil
| | - Deboraci B Prates
- Laboratório de Inflamação e Biomarcadores, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, Brazil.,Departamento de Biomorfologia, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| |
Collapse
|
29
|
Liu YP, Li L, Xu L, Dai EN, Chen WD. Cantharidin suppresses cell growth and migration, and activates autophagy in human non-small cell lung cancer cells. Oncol Lett 2018; 15:6527-6532. [PMID: 29731854 DOI: 10.3892/ol.2018.8141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022] Open
Abstract
Cantharidin (CTD), a component of Mylabris (blister beetle), is a traditional Chinese medicine that exerts an anticancer effect in multiple types of cancer cells. The aim of the present study was to investigate whether CTD exhibited anti-metastatic and inhibitory cell proliferation effects against human non-small cell lung cancer (NSCLC) A549 cells, and the possible underlying mechanism by which this occurs. The results of the present study demonstrated that CTD arrested proliferation, suppressed invasion and migration and induced apoptosis in A549 cells in vitro. Alterations of apoptosis-associated protein levels, including B-cell lymphoma-2 (Bcl-2), Bcl-associated X (Bax) and active caspase-3, were detected. Furthermore, the present study demonstrated that CTD activated autophagy through downregulation of p62 expression and upregulation of microtubule-associated proteins 1A/1B light chain 3B and Beclin-1 expression. Additionally, western blot analysis identified that CTD inhibited the phosphatidylinositol 3-kinase (PI3K)/RAC serine/threonine protein kinase (Akt)/mechanistic target of rapamycin (mTOR) signaling pathway in NSCLC, demonstrating that the levels of phosphorylated (p-)Akt, p-mTOR, phosphorylated ribosomal p70S6 protein kinase (p-p70-S6K) and cyclin D1 were significantly decreased following treatment with CTD. In conclusion, the results of the present study indicated that CTD impeded cell growth and migration by inhibiting PI3K/Akt/mTOR signaling in NSCLC, and promoted autophagy and apoptosis. CTD exhibited anticancer activity against NSCLC in vitro, revealing it as a potential candidate for the treatment of NSCLC.
Collapse
Affiliation(s)
- Yan-Peng Liu
- Department of Internal Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Ling Li
- Department of Surgery, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong 250031, P.R. China
| | - Liang Xu
- Department of Surgery, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong 250031, P.R. China
| | - E-Nuo Dai
- Department of Surgery, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong 250031, P.R. China
| | - Wei-Da Chen
- Department of Geriatric Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
30
|
Iba T, Levy JH. Inflammation and thrombosis: roles of neutrophils, platelets and endothelial cells and their interactions in thrombus formation during sepsis. J Thromb Haemost 2018; 16:231-241. [PMID: 29193703 DOI: 10.1111/jth.13911] [Citation(s) in RCA: 307] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Indexed: 12/12/2022]
Abstract
The inflammatory response and the activation of coagulation are two important responses in a host's defense against infection. These mechanisms do not work independently, but cooperate in a complex and synchronous manner. Recent research has also shed light on the critical role of thrombus formation, which prevents the dissemination of microorganisms. The cellular components of blood vessels, i.e. leukocytes, platelets, erythrocytes, and vascular endothelial cells, play significant roles in the development of thrombi in combination with activation of the coagulation system. In addition to the cellular components, alarmins such as histones and high-mobility group box 1, microparticles and secreted granule proteins are all important for clot formation. In this summary, we review the pathophysiology of sepsis-induced coagulopathy and the role of cellular components and critical factors released from damaged cells. In addition, we review important therapeutic approaches that have been developed, are under investigation and are currently available in certain countries, including antithrombin, recombinant thrombomodulin, anti-Toll-like receptor 4 therapy, anti-damage associated molecular pattern therapy, and hemoadsorption with a polymyxin B-immobilized fiber column.
Collapse
Affiliation(s)
- T Iba
- Department of Emergency and Disaster Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - J H Levy
- Department of Anesthesiology and Surgery, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
31
|
Necroptosis and neutrophil-associated disorders. Cell Death Dis 2018; 9:111. [PMID: 29371616 PMCID: PMC5833577 DOI: 10.1038/s41419-017-0058-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 07/13/2017] [Accepted: 07/16/2017] [Indexed: 12/11/2022]
Abstract
Necroptosis is a form of regulated necrosis and is dependent on a signaling pathway involving receptor interacting protein kinase-3 (RIPK3) and mixed lineage kinase domain-like protein (MLKL). Necroptosis is considered to have important functions in inflammation and, based on studies with animal disease models, is believed likely to be involved in the pathogenesis of many human inflammatory diseases. In neutrophils, necroptosis has recently been reported to be triggered by tumor necrosis factor (TNF) stimulation, ligation of adhesion receptors, exposure to monosodium urate (MSU) crystals, or phagocytosis of Staphylococcus aureus (S. aureus). Because neutrophils are involved in many kinds of tissue inflammation and disease, neutrophil necroptosis probably plays a vital role in such processes. Dissecting the signaling pathway of neutrophil necroptotic death may help to identify novel drug targets for inflammatory or autoimmune diseases. In this review, we discuss different mechanisms which regulate neutrophil necroptosis and are thus potentially important in neutrophil-associated disorders.
Collapse
|
32
|
Aulakh GK. Neutrophils in the lung: “the first responders”. Cell Tissue Res 2017; 371:577-588. [DOI: 10.1007/s00441-017-2748-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 11/21/2017] [Indexed: 12/27/2022]
|
33
|
Kono M, Saigo K, Yamamoto S, Shirai K, Iwamoto S, Uematsu T, Takahashi T, Imoto S, Hashimoto M, Minami Y, Wada A, Takenokuchi M, Kawano S. Iron-chelating agent, deferasirox, inhibits neutrophil activation and extracellular trap formation. Clin Exp Pharmacol Physiol 2017; 43:915-20. [PMID: 27333499 DOI: 10.1111/1440-1681.12612] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 06/19/2016] [Accepted: 06/20/2016] [Indexed: 01/19/2023]
Abstract
Iron-chelating agents, which are frequently prescribed to transfusion-dependent patients, have various useful biological effects in addition to chelation. Reactive oxygen species (ROS) produced by neutrophils can cause pulmonary endothelial cell damage, which can lead to acute lung injury (ALI). We previously reported that deferasirox (DFS), an iron-chelating agent, inhibits phorbol myristate acetate (PMA) or formyl-methionyl-leucyl-phenylalanine (fMLP)-induced ROS production in neutrophils, in vitro. Here, we investigate whether DFS inhibits vacuolization in neutrophils and neutrophil extracellular trap (NET) formation. Human neutrophils were incubated with DFS and stimulated with PMA or fMLP. Human neutrophils were separated from heparinized peripheral blood using density gradient centrifugation, and subsequently incubated with DFS. After 10 minutes, neutrophils were stimulated by PMA or fMLP. Vacuole formation was observed by electron microscopy. For observing NET formations using microscopes, immunohistological analyses using citrullinated histone H3 and myeloperoxidase antibodies, and SYTOX Green (an impermeable DNA detection dye) staining, were conducted. NET formation was measured as the quantity of double-stranded DNA (dsDNA), using the AccuBlue Broad Range dsDNA Quantitation Kit. DFS (50 μmol/L) inhibited vacuole formation in the cytoplasm and NET formation. Additionally, 5-100 μmol/L concentration of DFS inhibited the release of dsDNA in a dose-independent manner. We demonstrate that DFS inhibits not only ROS production but also vacuolization and NET formation in neutrophils. These results suggest the possibility of protective effects of DFS against NET-related adverse effects, including ALI and thrombosis.
Collapse
Affiliation(s)
- Mari Kono
- Scientific Research, Scientific Affairs, Sysmex Corporation, Kobe, Japan
| | - Katsuyasu Saigo
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| | - Shiori Yamamoto
- Scientific Research, Scientific Affairs, Sysmex Corporation, Kobe, Japan
| | - Kohei Shirai
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| | - Shuta Iwamoto
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| | - Tomoko Uematsu
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| | | | - Shion Imoto
- Department of Health Science, Kobe Tokiwa University, Kobe, Japan
| | | | | | - Atsushi Wada
- Scientific Research, Scientific Affairs, Sysmex Corporation, Kobe, Japan
| | - Mariko Takenokuchi
- Faculty of Pharmacological Sciences, Himeji Dokkyo University, Himeji, Japan
| | | |
Collapse
|
34
|
Germic N, Stojkov D, Oberson K, Yousefi S, Simon HU. Neither eosinophils nor neutrophils require ATG5-dependent autophagy for extracellular DNA trap formation. Immunology 2017; 152:517-525. [PMID: 28703297 DOI: 10.1111/imm.12790] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/06/2017] [Accepted: 07/06/2017] [Indexed: 12/18/2022] Open
Abstract
The importance of extracellular traps (ETs) in innate immunity is well established, but the molecular mechanisms responsible for their formation remain unclear and in scientific dispute. ETs have been defined as extracellular DNA scaffolds associated with the granule proteins of eosinophils or neutrophils. They are capable of killing bacteria extracellularly. Based mainly on results with phosphoinositide 3-kinase (PI3K) inhibitors such as 3-methyladenine (3-MA) and wortmannin, which are commonly used to inhibit autophagy, several groups have reported that autophagy is required for neutrophil extracellular trap (NET) formation. We decided to investigate this apparent dependence on autophagy for ET release and generated genetically modified mice that lack, specifically in eosinophils or neutrophils, autophagy-related 5 (Atg5), a gene encoding a protein essential for autophagosome formation. Interestingly, neither eosinophils nor neutrophils from Atg5-deficient mice exhibited abnormalities in ET formation upon physiological activation or exposure to low concentrations of PMA, although we could confirm that human and mouse eosinophils and neutrophils, after pre-treatment with inhibitors of class III PI3K, show a block both in reactive oxygen species (ROS) production and in ET formation. The so-called late autophagy inhibitors bafilomycin A1 and chloroquine, on the other hand, were without effect. These data indicate that ET formation occurs independently of autophagy and that the inhibition of ROS production and ET formation in the presence of 3-MA and wortmannin is probably owing to their additional ability to block the class I PI3Ks, which are involved in signalling cascades initiated by triggers of ET formation.
Collapse
Affiliation(s)
- Nina Germic
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Darko Stojkov
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Kevin Oberson
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| |
Collapse
|
35
|
Qian M, Fang X, Wang X. Autophagy and inflammation. Clin Transl Med 2017; 6:24. [PMID: 28748360 PMCID: PMC5529308 DOI: 10.1186/s40169-017-0154-5] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 07/18/2017] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a homeostatic mechanism involved in the disposal of damaged organelles, denatured proteins as well as invaded pathogens through a lysosomal degradation pathway. Recently, increasing evidences have demonstrated its role in both innate and adaptive immunity, and thereby influence the pathogenesis of inflammatory diseases. The detection of autophagy machinery facilitated the measurement of autophagy during physiological and pathophysiological processes. Autophagy plays critical roles in inflammation through influencing the development, homeostasis and survival of inflammatory cells, including macrophages, neutrophils and lymphocytes; effecting the transcription, processing and secretion of a number of cytokines, as well as being regulated by cytokines. Recently, autophagy-dependent mechanisms have been studied in the pathogenesis of several inflammatory diseases, including infectious diseases, Crohn’s disease, cystic fibrosis, pulmonary hypertension, chronic obstructive pulmonary diseases and so on. These studies suggested that modulation of autophagy might lead to therapeutic interventions for diseases associated with inflammation. Here we highlight recent advances in investigating the roles of autophagy in inflammation as well as inflammatory diseases.
Collapse
Affiliation(s)
- Mengjia Qian
- Zhongshan Hospital Institute of Clinical Science, Shanghai Institute of Clinical Bioinformatics, Fudan University Medical School, Shanghai, China
| | - Xiaocong Fang
- Zhongshan Hospital Institute of Clinical Science, Shanghai Institute of Clinical Bioinformatics, Fudan University Medical School, Shanghai, China
| | - Xiangdong Wang
- Zhongshan Hospital Institute of Clinical Science, Shanghai Institute of Clinical Bioinformatics, Fudan University Medical School, Shanghai, China.
| |
Collapse
|
36
|
Schneider C, Wicki S, Graeter S, Timcheva TM, Keller CW, Quast I, Leontyev D, Djoumerska-Alexieva IK, Käsermann F, Jakob SM, Dimitrova PA, Branch DR, Cummings RD, Lünemann JD, Kaufmann T, Simon HU, von Gunten S. IVIG regulates the survival of human but not mouse neutrophils. Sci Rep 2017; 7:1296. [PMID: 28465620 PMCID: PMC5430961 DOI: 10.1038/s41598-017-01404-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 03/30/2017] [Indexed: 12/12/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) are purified IgG preparations made from the pooled plasma from thousands of healthy donors and are being tested in preclinical mouse models. Inherent challenges, however, are the pluripotency of IVIG and its xenogeneicity in animals. IVIG can alter the viability of human neutrophils via agonistic antibodies to Fas and Siglec-9. In this study, we compared the effects of IVIG on human and mouse neutrophils using different death assays. Different commercial IVIG preparations similarly induced cytokine-dependent death in human neutrophils, whereas they had no effects on the survival of either peripheral blood or bone marrow neutrophils from C57BL/6 or BALB/c mice. F(ab’)2 but not Fc fragments of IVIG induced death of human neutrophils, whereas neither of these IVIG fragments, nor agonistic monoclonal antibodies to human Fas or Siglec-9 affected the viability of mouse neutrophils. Pooled mouse IgG, which exhibited a different immunoprofile compared to IVIG, also had no effect on mouse cells. Together, these observations demonstrate that effects of IVIG on neutrophil survival are not adequately reflected in current mouse models, despite the key role of these cells in human inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
| | - Simone Wicki
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stefanie Graeter
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | | - Christian W Keller
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Isaak Quast
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Danila Leontyev
- Department of Medicine, University of Toronto and Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Iglika K Djoumerska-Alexieva
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Stephan M Jakob
- Department of Intensive Care Medicine, University Hospital Bern (Inselspital), University of Bern, Bern, Switzerland
| | - Petya A Dimitrova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Donald R Branch
- Department of Medicine, University of Toronto and Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jan D Lünemann
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | |
Collapse
|
37
|
Radonjic-Hoesli S, Wang X, de Graauw E, Stoeckle C, Styp-Rekowska B, Hlushchuk R, Simon D, Spaeth PJ, Yousefi S, Simon HU. Adhesion-induced eosinophil cytolysis requires the receptor-interacting protein kinase 3 (RIPK3)-mixed lineage kinase-like (MLKL) signaling pathway, which is counterregulated by autophagy. J Allergy Clin Immunol 2017; 140:1632-1642. [PMID: 28412393 DOI: 10.1016/j.jaci.2017.01.044] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/22/2016] [Accepted: 01/12/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND Eosinophils are a subset of granulocytes that can be involved in the pathogenesis of different diseases, including allergy. Their effector functions are closely linked to their cytotoxic granule proteins. Release takes place through several different mechanisms, one of which is cytolysis, which is associated with release of intact granules, so-called clusters of free eosinophil granules. The mechanism underlying this activation-induced form of cell death in eosinophils has remained unclear. OBJECTIVE We aimed to elucidate the molecular mechanism of eosinophil cytolysis. METHODS Isolated blood eosinophils were incubated on glass coverslips coated with intravenous immunoglobulin and inactive complement component 3b. A morphologic characterization of the distinct stages of the proposed cascade was addressed by means of time-lapse automated fluorescence microscopy, electron microscopy, and immunohistochemistry. Experiments with pharmacologic inhibitors were performed to elucidate the sequence of events within the cascade. Tissue samples of patients with eosinophilic skin diseases or eosinophilic esophagitis were used for in vivo analyses. RESULTS After eosinophil adhesion, we observed reactive oxygen species production, early degranulation, and granule fusion processes, leading to a distinct morphology exhibiting cytoplasmic vacuolization and, finally, cytolysis. Using a pharmacologic approach, we demonstrate the presence of a receptor-interacting protein kinase 3 (RIPK3)-mixed lineage kinase-like (MLKL) signaling pathway in eosinophils, which, after its activation, leads to the production of high levels of reactive oxygen species in a p38 mitogen-activated protein kinase and phosphatidylinositol 3'-kinase-dependent manner. All these steps are required for cytoplasmic vacuolization and subsequent cytolysis to occur. Interestingly, triggering cytolysis is associated with an induction of autophagy in eosinophils, and additional stimulation of autophagy by means of pharmacologic inhibition of the mechanistic target of rapamycin counterregulates cell death. Moreover, MLKL phosphorylation, cytoplasmic vacuolization, and cytolysis were observed in eosinophils under in vivo inflammatory conditions. CONCLUSION We report that adhesion-induced eosinophil cytolysis takes place through RIPK3-MLKL-dependent necroptosis, which can be counterregulated by autophagy.
Collapse
Affiliation(s)
- Susanne Radonjic-Hoesli
- Institute of Pharmacology, University of Bern, Bern, Switzerland; Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xiaoliang Wang
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | | | | | | | | - Dagmar Simon
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Peter J Spaeth
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Shida Yousefi
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland.
| |
Collapse
|
38
|
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
|
39
|
Lavie L, Dyugovskaya L, Polyakov A, Rogovoy O, Leder E. Development and Identification of a Novel Subpopulation of Human Neutrophil-derived Giant Phagocytes In Vitro. J Vis Exp 2017:54826. [PMID: 28190059 PMCID: PMC5352295 DOI: 10.3791/54826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Neutrophils (PMN) are best known for their phagocytic functions against invading pathogens and microorganisms. They have the shortest half-life amongst leukocytes and in their non-activated state are constitutively committed to apoptosis. When recruited to inflammatory sites to resolve inflammation, they produce an array of cytotoxic molecules with potent antimicrobial killing. Yet, when these powerful cytotoxic molecules are released in an uncontrolled manner they can damage surrounding tissues. In recent years however, neutrophil versatility is increasingly evidenced, by demonstrating plasticity and immunoregulatory functions. We have recently identified a new neutrophil-derived subpopulation, which develops spontaneously in standard culture conditions without the addition of cytokines/growth factors such as granulocyte colony-stimulating factor (GM-CSF)/interleukin (IL)-4. Their phagocytic abilities of neutrophil remnants largely contribute to increase their size immensely; therefore they were termed giant phagocytes (Gϕ). Unlike neutrophils, Gϕ are long lived in culture. They express the cluster of differentiation (CD) neutrophil markers CD66b/CD63/CD15/CD11b/myeloperoxidase (MPO)/neutrophil elastase (NE), and are devoid of the monocytic lineage markers CD14/CD16/CD163 and the dendritic CD1c/CD141 markers. They also take-up latex and zymosan, and respond by oxidative burst to stimulation with opsonized-zymosan and PMA. Gϕ also express the scavenger receptors CD68/CD36, and unlike neutrophils, internalize oxidized-low density lipoprotein (oxLDL). Moreover, unlike fresh neutrophils, or cultured monocytes, they respond to oxLDL uptake by increased reactive oxygen species (ROS) production. Additionally, these phagocytes contain microtubule-associated protein-1 light chain 3B (LC3B) coated vacuoles, indicating the activation of autophagy. Using specific inhibitors it is evident that both phagocytosis and autophagy are prerequisites for their development and likely NADPH oxidase dependent ROS. We describe here a method for the preparation of this new subpopulation of long-lived, neutrophil-derived phagocytic cells in culture, their identification and their currently known characteristics. This protocol is essential for obtaining and characterizing Gϕ in order to further investigate their significance and functions.
Collapse
Affiliation(s)
- Lena Lavie
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Insitute of Technology;
| | - Larissa Dyugovskaya
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Insitute of Technology
| | - Andrey Polyakov
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Insitute of Technology
| | - Oksana Rogovoy
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Insitute of Technology
| | - Eva Leder
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Insitute of Technology
| |
Collapse
|
40
|
Dos Anjos DO, Sobral Alves ES, Gonçalves VT, Fontes SS, Nogueira ML, Suarez-Fontes AM, Neves da Costa JB, Rios-Santos F, Vannier-Santos MA. Effects of a novel β-lapachone derivative on Trypanosoma cruzi: Parasite death involving apoptosis, autophagy and necrosis. Int J Parasitol Drugs Drug Resist 2016; 6:207-219. [PMID: 27770751 PMCID: PMC5078628 DOI: 10.1016/j.ijpddr.2016.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/07/2016] [Accepted: 10/10/2016] [Indexed: 12/18/2022]
Abstract
Natural products comprise valuable sources for new antiparasitic drugs. Here we tested the effects of a novel β-lapachone derivative on Trypanosoma cruzi parasite survival and proliferation and used microscopy and cytometry techniques to approach the mechanism(s) underlying parasite death. The selectivity index determination indicate that the compound trypanocidal activity was over ten-fold more cytotoxic to epimastigotes than to macrophages or splenocytes. Scanning electron microscopy analysis revealed that the R72 β-lapachone derivative affected the T. cruzi morphology and surface topography. General plasma membrane waving and blebbing particularly on the cytostome region were observed in the R72-treated parasites. Transmission electron microscopy observations confirmed the surface damage at the cytostome opening vicinity. We also observed ultrastructural evidence of the autophagic mechanism termed macroautophagy. Some of the autophagosomes involved large portions of the parasite cytoplasm and their fusion/confluence may lead to necrotic parasite death. The remarkably enhanced frequency of autophagy triggering was confirmed by quantitating monodansylcadaverine labeling. Some cells displayed evidence of chromatin pycnosis and nuclear fragmentation were detected. This latter phenomenon was also indicated by DAPI staining of R72-treated cells. The apoptotis induction was suggested to take place in circa one-third of the parasites assessed by annexin V labeling measured by flow cytometry. TUNEL staining corroborated the apoptosis induction. Propidium iodide labeling indicate that at least 10% of the R72-treated parasites suffered necrosis within 24 h. The present data indicate that the β-lapachone derivative R72 selectively triggers T. cruzi cell death, involving both apoptosis and autophagy-induced necrosis.
Collapse
Affiliation(s)
- Danielle Oliveira Dos Anjos
- Lab. Biologia Parasitária, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz - FIOCRUZ, Brazil; Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz UESC, Brazil
| | | | | | - Sheila Suarez Fontes
- Lab. Biologia Parasitária, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz - FIOCRUZ, Brazil
| | - Mateus Lima Nogueira
- Lab. Biologia Parasitária, Instituto Gonçalo Moniz, Fundação Oswaldo Cruz - FIOCRUZ, Brazil
| | | | | | | | | |
Collapse
|
41
|
Wang X, He Z, Liu H, Yousefi S, Simon HU. Neutrophil Necroptosis Is Triggered by Ligation of Adhesion Molecules following GM-CSF Priming. THE JOURNAL OF IMMUNOLOGY 2016; 197:4090-4100. [DOI: 10.4049/jimmunol.1600051] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 09/12/2016] [Indexed: 01/08/2023]
|
42
|
Activation of Adenosine 2A receptor inhibits neutrophil apoptosis in an autophagy-dependent manner in mice with systemic inflammatory response syndrome. Sci Rep 2016; 6:33614. [PMID: 27647162 PMCID: PMC5028892 DOI: 10.1038/srep33614] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/30/2016] [Indexed: 01/13/2023] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is an overwhelming whole body inflammation caused by infectious diseases or sterile insults. Neutrophils are the dominant participants during inflammation, and their survival and death determine the initiation as well as resolution of SIRS. Apoptosis and autophagy are two fundamental cellular processes that modulating cell fate, but their correlation and regulators in neutrophils under SIRS condition have not been elucidated. In this study, we demonstrated that high dose of LPS induced both apoptosis and autophagy of neutrophils in a mouse SIRS model and LPS-stimulated neutrophils in vitro. Moreover, we found that the adenosine 2A receptor (A2AR), a known anti-inflammatory G protein-coupled receptor (GPCR), could inhibit LPS-induced neutrophil apoptosis by suppressing the LPS-induced autophagy. Activation of A2AR suppressed LPS-induced autophagy by inhibiting the ROS-JNK pathway as well as promoting GPCR βϒ subunit–AKT signaling. The A2AR-inhibited autophagy suppressed apoptosis of neutrophils by blocking caspase8, caspase3 and PARP signaling. These findings not only increase our understandings of neutrophils’ fate and function in response to systemic inflammation, but also identify a novel anti-inflammatory role of A2AR in modulating neutrophils’ survival during inflammation.
Collapse
|
43
|
Adhauliya N, Kalappanavar AN, Ali IM, Annigeri RG. Autophagy: A boon or bane in oral cancer. Oral Oncol 2016; 61:120-6. [PMID: 27688114 DOI: 10.1016/j.oraloncology.2016.09.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 02/06/2023]
Abstract
Autophagy is a catabolic process involving cellular recycling and is believed to play a distinct role in cell survival especially when exposed to stressors, rendering it comparable to the elixir sustaining life. It plays a significant role in various conditions like cancers, neuropathies, heart diseases, auto-immune diseases, etc. Its role in tumorigenesis and cancer therapeutics is worth exploring. Autophagy is believed to help in survival and longevity of cancer cells by buffering metabolic stress. Inhibition of autophagy in an environment of nutrient deprivation leads to cell death. Autophagy is also seen to facilitate metastasizing tumor cells in surviving the conditions of metabolic deprivation and in recovery when conditions turn favorable. Many current cancer therapies tend to inflict metabolic stress, thus autophagy inhibitors may be useful in cancer treatment. As per the adage, "excess of anything is bad", the autophagy promoters can also be exploited as beneficial tools in the fight against cancer. Another method for tumor-cell elimination can be by inducing autophagic cell death through over-stimulation. Oral cancers are becoming a leading cause of deaths worldwide. Much remains to be explored about the role autophagy plays in progression of head and neck cancers, so as to harness it in the therapeutics of these cancers. Research on autophagy is still in its infancy. There are knowledge gaps in understanding this complex process. But there is no doubt that understanding exact mechanism behind autophagy will open up new avenues in cancer therapeutics and even prevention.
Collapse
Affiliation(s)
- Namrata Adhauliya
- Department of Oral Medicine and Radiology, College of Dental Sciences, Davangere, Karnataka, India.
| | - Anupama N Kalappanavar
- Department of Oral Medicine and Radiology, College of Dental Sciences, Davangere, Karnataka, India
| | - I M Ali
- Department of Oral Medicine and Radiology, College of Dental Sciences, Davangere, Karnataka, India
| | - Rajeshwari G Annigeri
- Department of Oral Medicine and Radiology, College of Dental Sciences, Davangere, Karnataka, India
| |
Collapse
|
44
|
Yousefi S, Simon HU. NETosis - Does It Really Represent Nature's "Suicide Bomber"? Front Immunol 2016; 7:328. [PMID: 27617015 PMCID: PMC4999959 DOI: 10.3389/fimmu.2016.00328] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 08/16/2016] [Indexed: 12/31/2022] Open
Affiliation(s)
- Shida Yousefi
- Institute of Pharmacology, University of Bern , Bern , Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern , Bern , Switzerland
| |
Collapse
|
45
|
Fullerton JN, Gilroy DW. Resolution of inflammation: a new therapeutic frontier. Nat Rev Drug Discov 2016; 15:551-67. [PMID: 27020098 DOI: 10.1038/nrd.2016.39] [Citation(s) in RCA: 618] [Impact Index Per Article: 68.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dysregulated inflammation is a central pathological process in diverse disease states. Traditionally, therapeutic approaches have sought to modulate the pro- or anti-inflammatory limbs of inflammation, with mixed success. However, insight into the pathways by which inflammation is resolved has highlighted novel opportunities to pharmacologically manipulate these processes - a strategy that might represent a complementary (and perhaps even superior) therapeutic approach. This Review discusses the state of the art in the biology of resolution of inflammation, highlighting the opportunities and challenges for translational research in this field.
Collapse
Affiliation(s)
- James N Fullerton
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street, University College London, London WC1E 6JJ, UK
| | - Derek W Gilroy
- Centre for Clinical Pharmacology and Therapeutics, Division of Medicine, 5 University Street, University College London, London WC1E 6JJ, UK
| |
Collapse
|
46
|
Dyugovskaya L, Berger S, Polyakov A, Lavie P, Lavie L. Intermittent Hypoxia Affects the Spontaneous Differentiation In Vitro of Human Neutrophils into Long-Lived Giant Phagocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:9636937. [PMID: 26635914 PMCID: PMC4655297 DOI: 10.1155/2016/9636937] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/01/2015] [Indexed: 11/17/2022]
Abstract
Previously we identified, for the first time, a new small-size subset of neutrophil-derived giant phagocytes (Gϕ) which spontaneously develop in vitro without additional growth factors or cytokines. Gϕ are CD66b(+)/CD63(+)/MPO(+)/LC3B(+) and are characterized by extended lifespan, large phagolysosomes, active phagocytosis, and reactive oxygen species (ROS) production, and autophagy largely controls their formation. Hypoxia, and particularly hypoxia/reoxygenation, is a prominent feature of many pathological processes. Herein we investigated Gϕ formation by applying various hypoxic conditions. Chronic intermittent hypoxia (IH) (29 cycles/day for 5 days) completely abolished Gϕ formation, while acute IH had dose-dependent effects. Exposure to 24 h (56 IH cycles) decreased their size, yield, phagocytic ability, autophagy, mitophagy, and gp91-phox/p22-phox expression, whereas under 24 h sustained hypoxia (SH) the size and expression of LC3B and gp91-phox/p22-phox resembled Gϕ formed in normoxia. Diphenyl iodide (DPI), a NADPH oxidase inhibitor, as well as the PI3K/Akt and autophagy inhibitor LY294002 abolished Gϕ formation at all oxygen conditions. However, the potent antioxidant, N-acetylcysteine (NAC) abrogated the effects of IH by inducing large CD66b(+)/LC3B(+) Gϕ and increased both NADPH oxidase expression and phagocytosis. These findings suggest that NADPH oxidase, autophagy, and the PI3K/Akt pathway are involved in Gϕ development.
Collapse
Affiliation(s)
- Larissa Dyugovskaya
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 31096 Haifa, Israel
| | - Slava Berger
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 31096 Haifa, Israel
| | - Andrey Polyakov
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 31096 Haifa, Israel
| | - Peretz Lavie
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 31096 Haifa, Israel
| | - Lena Lavie
- The Lloyd Rigler Sleep Apnea Research Laboratory, Unit of Anatomy and Cell Biology, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 31096 Haifa, Israel
| |
Collapse
|
47
|
Yousefi S, Morshed M, Amini P, Stojkov D, Simon D, von Gunten S, Kaufmann T, Simon HU. Basophils exhibit antibacterial activity through extracellular trap formation. Allergy 2015; 70:1184-8. [PMID: 26043360 DOI: 10.1111/all.12662] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2015] [Indexed: 01/21/2023]
Abstract
Basophils are primarily associated with immunomodulatory functions in allergic diseases and parasitic infections. Recently, it has been demonstrated that both activated human and mouse basophils can form extracellular DNA traps (BETs) containing mitochondrial DNA and granule proteins. In this report, we provide evidence that, in spite of an apparent lack of phagocytic activity, basophils can kill bacteria through BET formation.
Collapse
Affiliation(s)
- S. Yousefi
- Institute of Pharmacology; University of Bern; Bern Switzerland
| | - M. Morshed
- Institute of Pharmacology; University of Bern; Bern Switzerland
| | - P. Amini
- Institute of Pharmacology; University of Bern; Bern Switzerland
| | - D. Stojkov
- Institute of Pharmacology; University of Bern; Bern Switzerland
| | - D. Simon
- Department of Dermatology; Inselspital, Bern University Hospital; Bern Switzerland
| | - S. von Gunten
- Institute of Pharmacology; University of Bern; Bern Switzerland
| | - T. Kaufmann
- Institute of Pharmacology; University of Bern; Bern Switzerland
| | - H.-U. Simon
- Institute of Pharmacology; University of Bern; Bern Switzerland
| |
Collapse
|
48
|
Yang X, Yu DD, Yan F, Jing YY, Han ZP, Sun K, Liang L, Hou J, Wei LX. The role of autophagy induced by tumor microenvironment in different cells and stages of cancer. Cell Biosci 2015; 5:14. [PMID: 25844158 PMCID: PMC4384293 DOI: 10.1186/s13578-015-0005-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 03/13/2015] [Indexed: 12/13/2022] Open
Abstract
Development of a tumor is a very complex process, and invasion and metastasis of malignant tumors are hallmarks and are difficult problems to overcome. The tumor microenvironment plays an important role in controlling tumor fate and autophagy induced by the tumor microenvironment is attracting more and more attention. Autophagy can be induced by several stressors in the tumor microenvironment and autophagy modifies the tumor microenvironment, too. Autophagy has dual roles in tumor growth. In this review, we discussed the interaction between autophagy and the tumor microenvironment and the paradoxical roles of autophagy on tumor growth at different stages of tumor development.
Collapse
Affiliation(s)
- Xue Yang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Dan-Dan Yu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Fei Yan
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Ying-Ying Jing
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Zhi-Peng Han
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Kai Sun
- Central laboratory, Ren Ji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Lei Liang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| | - Jing Hou
- Department of Pharmacy, Chang Hai Hospital, The Second Military Medical University, Shanghai, China
| | - Li-Xin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, 225 Changhai Road, 200438 Shanghai, China
| |
Collapse
|
49
|
Wehrli M, Cortinas-Elizondo F, Hlushchuk R, Daudel F, Villiger PM, Miescher S, Zuercher AW, Djonov V, Simon HU, von Gunten S. Human IgA Fc Receptor FcαRI (CD89) Triggers Different Forms of Neutrophil Death Depending on the Inflammatory Microenvironment. THE JOURNAL OF IMMUNOLOGY 2014; 193:5649-59. [DOI: 10.4049/jimmunol.1400028] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
50
|
Romero V, Fert-Bober J, Nigrovic PA, Darrah E, Haque UJ, Lee DM, van Eyk J, Rosen A, Andrade F. Immune-mediated pore-forming pathways induce cellular hypercitrullination and generate citrullinated autoantigens in rheumatoid arthritis. Sci Transl Med 2014; 5:209ra150. [PMID: 24174326 DOI: 10.1126/scitranslmed.3006869] [Citation(s) in RCA: 171] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Autoantibodies to citrullinated protein antigens are specific markers of rheumatoid arthritis (RA). Although protein citrullination can be activated by numerous stimuli in cells, it remains unclear which of these produce the prominent citrullinated autoantigens targeted in RA. In these studies, we show that RA synovial fluid cells have an unusual pattern of citrullination with marked citrullination of proteins across the broad range of molecular weights, which we term cellular hypercitrullination. Although histone citrullination is a common event during neutrophil activation and death induced by different pathways including apoptosis, NETosis, and necroptosis/autophagy, hypercitrullination is not induced by these stimuli. However, marked hypercitrullination is induced by two immune-mediated membranolytic pathways, mediated by perforin and the membrane attack complex (MAC), which are active in the RA joint and of importance in RA pathogenesis. We further demonstrate that perforin and MAC activity on neutrophils generate the profile of citrullinated autoantigens characteristic of RA. These data suggest that activation of peptidylarginine deiminases during complement and perforin activity may be at the core of citrullinated autoantigen production in RA. These pathways may be amenable to monitoring and therapeutic modulation.
Collapse
Affiliation(s)
- Violeta Romero
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | |
Collapse
|