1
|
Lecomte K, Toniolo A, Hoste E. Cell death as an architect of adult skin stem cell niches. Cell Death Differ 2024; 31:957-969. [PMID: 38649745 PMCID: PMC11303411 DOI: 10.1038/s41418-024-01297-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Our skin provides a physical and immunological barrier against dehydration and environmental insults ranging from microbial attacks, toxins and UV irradiation to wounding. Proper functioning of the skin barrier largely depends on the interplay between keratinocytes- the epithelial cells of the skin- and immune cells. Two spatially distinct populations of keratinocyte stem cells (SCs) maintain the epidermal barrier function and the hair follicle. These SCs are inherently long-lived, but cell death can occur within their niches and impacts their functionality. The default cell death programme in skin is apoptosis, an orderly and non-inflammatory suicide programme. However, recent findings are shedding light on the significance of various modes of regulated necrotic cell death, which are lytic and can provoke inflammation within the local skin environment. While the presence of dying cells was generally regarded as a mere consequence of inflammation, findings in various human dermatological conditions and experimental mouse models of aberrant cell death control demonstrated that cell death programmes in keratinocytes (KCs) can drive skin inflammation and even tumour initiation. When cells die, they need to be removed by phagocytosis and KCs can function as non-professional phagocytes of apoptotic cells with important implications for their SC capacities. It is becoming apparent that in conditions of heightened SC activity, distinct cell death modalities differentially impact the different skin SC populations in their local niches. Here, we describe how regulated cell death modalities functionally affect epidermal SC niches along with their relevance to injury repair, inflammatory skin disorders and cancer.
Collapse
Affiliation(s)
- Kim Lecomte
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Annagiada Toniolo
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Esther Hoste
- VIB Center for Inflammation Research, 9052, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium.
| |
Collapse
|
2
|
Kamprom W, Tangporncharoen R, Vongthaiwan N, Tragoonlugkana P, Phetfong J, Pruksapong C, Supokawej A. Enhanced potent immunosuppression of intracellular adipose tissue-derived stem cell extract by priming with three-dimensional spheroid formation. Sci Rep 2024; 14:9084. [PMID: 38643332 PMCID: PMC11032398 DOI: 10.1038/s41598-024-59910-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/16/2024] [Indexed: 04/22/2024] Open
Abstract
Immunomodulatory properties of mesenchymal stem cells are widely studied, supporting the use of MSCs as cell-based therapy in immunological diseases. This study aims to generate cell-free MSC extract and improves their immunomodulatory potential. Intracellular extracts were prepared from adipose-derived stem cells (ADSC) spheroid via a freeze-thawing method. The immunomodulatory capacities of ADSC spheroid extracts were investigated in vitro, including lymphocyte proliferation, T regulatory cell expansion, and macrophage assays. A comparative study was conducted with ADSC monolayer extract. The key immunomodulatory mediators presented in ADSC extract were identified. The results revealed that ADSC spheroid extract could suppress lymphocyte activation while enhancing T regulatory cell expansion. Immunomodulatory molecules such as COX-2, TSG-6, and TGF-β1 were upregulated in ADSC priming via spheroid culture. Selective inhibition of COX-2 abrogates the effect of ADSC extract on inducing T regulatory cell expansion. Thus, ADSC spheroid extract gains high efficacy in regulating the immune responses which are associated in part by COX-2 generation. Furthermore, ADSC spheroid extract possessed a potent anti-inflammation by manipulation of TNF-α production from LPS-activated macrophage. Our current study has highlighted the opportunity of using cell-free extracts from adipose tissue-derived mesenchymal stem cells spheroid as novel immunomodulators for the treatment of immunological-associated diseases.
Collapse
Affiliation(s)
- Witchayapon Kamprom
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Rattanawan Tangporncharoen
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Nuttapoom Vongthaiwan
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Patcharapa Tragoonlugkana
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Jitrada Phetfong
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand
| | - Chatchai Pruksapong
- Department of Surgery, Phramongkutklao Hospital and College of Medicine, Bangkok, Thailand
| | - Aungkura Supokawej
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, 999 Phutthamonthon Sai 4, Salaya, Phutthamonthon, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
3
|
Glaubitz J, Asgarbeik S, Lange R, Mazloum H, Elsheikh H, Weiss FU, Sendler M. Immune response mechanisms in acute and chronic pancreatitis: strategies for therapeutic intervention. Front Immunol 2023; 14:1279539. [PMID: 37881430 PMCID: PMC10595029 DOI: 10.3389/fimmu.2023.1279539] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023] Open
Abstract
Acute pancreatitis (AP) is one of the most common inflammatory diseases of the gastrointestinal tract and a steady rising diagnosis for inpatient hospitalization. About one in four patients, who experience an episode of AP, will develop chronic pancreatitis (CP) over time. While the initiating causes of pancreatitis can be complex, they consistently elicit an immune response that significantly determines the severity and course of the disease. Overall, AP is associated with a significant mortality rate of 1-5%, which is caused by either an excessive pro-inflammation, or a strong compensatory inhibition of bacterial defense mechanisms which lead to a severe necrotizing form of pancreatitis. At the time-point of hospitalization the already initiated immune response is the only promising common therapeutic target to treat or prevent a severe disease course. However, the complexity of the immune response requires fine-balanced therapeutic intervention which in addition is limited by the fact that a significant proportion of patients is in danger of development or progress to recurrent and chronic disease. Based on the recent literature we survey the disease-relevant immune mechanisms and evaluate appropriate and promising therapeutic targets for the treatment of acute and chronic pancreatitis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Matthias Sendler
- Department of Medicine A, University Medicine, University of Greifswald, Greifswald, Germany
| |
Collapse
|
4
|
Abstract
Bronchiectasis is a final common pathway of a wide variety of underlying conditions including infectious, autoimmune, allergic, genetic and inflammatory conditions. Patients experience a chronic disease with variable clinical symptoms and course, but most experience cough, sputum production and recurrent exacerbations. Symptoms of bronchiectasis lead to poor quality of life and exacerbations are the major driver of morbidity and mortality. Patients are often chronically infected with bacteria with the most common being Pseudomonas aeruginosa and Haemophilus influenzae. Treatment of bronchiectasis includes standardised testing to identify the underlying cause with targeted treatment if immune deficiency, allergic bronchopulmonary aspergillosis or non-tuberculous mycobacterial infection, for example, are identified. Airway clearance is the mainstay of therapy for patients with symptoms of cough and sputum production. Frequently exacerbating patients may benefit from long term antibiotic or mucoactive therapies. Bronchiectasis is a heterogeneous disease and increasingly precision medicine approaches are advocated to target treatments most appropriately and to limit the emergence of antimicrobial resistance.
Collapse
Affiliation(s)
- Miguel Barbosa
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, Dundee, DD1 9SY, UK.
| |
Collapse
|
5
|
Ribeiro HAL, Scindia Y, Mehrad B, Laubenbacher R. COVID-19-associated pulmonary aspergillosis in immunocompetent patients: a virtual patient cohort study. J Math Biol 2023; 87:6. [PMID: 37306747 DOI: 10.1007/s00285-023-01940-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 05/10/2023] [Accepted: 05/21/2023] [Indexed: 06/13/2023]
Abstract
The opportunistic fungus Aspergillus fumigatus infects the lungs of immunocompromised hosts, including patients undergoing chemotherapy or organ transplantation. More recently however, immunocompetent patients with severe SARS-CoV2 have been reported to be affected by COVID-19 Associated Pulmonary Aspergillosis (CAPA), in the absence of the conventional risk factors for invasive aspergillosis. This paper explores the hypothesis that contributing causes are the destruction of the lung epithelium permitting colonization by opportunistic pathogens. At the same time, the exhaustion of the immune system, characterized by cytokine storms, apoptosis, and depletion of leukocytes may hinder the response to A. fumigatus infection. The combination of these factors may explain the onset of invasive aspergillosis in immunocompetent patients. We used a previously published computational model of the innate immune response to infection with Aspergillus fumigatus. Variation of model parameters was used to create a virtual patient population. A simulation study of this virtual patient population to test potential causes for co-infection in immunocompetent patients. The two most important factors determining the likelihood of CAPA were the inherent virulence of the fungus and the effectiveness of the neutrophil population, as measured by granule half-life and ability to kill fungal cells. Varying these parameters across the virtual patient population generated a realistic distribution of CAPA phenotypes observed in the literature. Computational models are an effective tool for hypothesis generation. Varying model parameters can be used to create a virtual patient population for identifying candidate mechanisms for phenomena observed in actual patient populations.
Collapse
Affiliation(s)
- Henrique A L Ribeiro
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, 32610, FL, USA
| | - Yogesh Scindia
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, 32610, FL, USA
| | - Borna Mehrad
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, 32610, FL, USA
| | - Reinhard Laubenbacher
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Florida, Gainesville, 32610, FL, USA.
| |
Collapse
|
6
|
Dvoriantchikova G, Adis E, Lypka K, Ivanov D. Various Forms of Programmed Cell Death Are Concurrently Activated in the Population of Retinal Ganglion Cells after Ischemia and Reperfusion. Int J Mol Sci 2023; 24:9892. [PMID: 37373037 DOI: 10.3390/ijms24129892] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Retinal ischemia-reperfusion (IR)-which ultimately results in retinal ganglion cell (RGC) death-is a common cause of visual impairment and blindness worldwide. IR results in various types of programmed cell death (PCD), which are of particular importance since they can be prevented by inhibiting the activity of their corresponding signaling cascades. To study the PCD pathways in ischemic RGCs, we used a mouse model of retinal IR and a variety of approaches including RNA-seq analysis, knockout animals, and animals treated with an iron chelator. In our RNA-seq analysis, we utilized RGCs isolated from retinas 24 h after IR. In ischemic RGCs, we found increased expression of many genes that regulate apoptosis, necroptosis, pyroptosis, oxytosis/ferroptosis, and parthanatos. Our data indicate that genetic ablation of death receptors protects RGCs from IR. We showed that the signaling cascades regulating ferrous iron (Fe2+) metabolism undergo significant changes in ischemic RGCs, leading to retinal damage after IR. This data suggests that the activation of death receptors and increased Fe2+ production in ischemic RGCs promote the simultaneous activation of apoptosis, necroptosis, pyroptosis, oxytosis/ferroptosis, and parthanatos pathways. Thus, a therapy is needed that concurrently regulates the activity of the multiple PCD pathways to reduce RGC death after IR.
Collapse
Affiliation(s)
- Galina Dvoriantchikova
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Emily Adis
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Karin Lypka
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dmitry Ivanov
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
7
|
Huang W, Kraynak CA, Bender EC, Farrar RP, Suggs LJ. Soluble components from mesenchymal stromal cell processing exert anti-inflammatory effects and facilitate ischemic muscle regeneration. Cytotherapy 2023; 25:375-386. [PMID: 36543717 PMCID: PMC10006307 DOI: 10.1016/j.jcyt.2022.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AIMS Skeletal muscle regeneration after severe damage is reliant on local stem cell proliferation and differentiation, processes that are tightly regulated by macrophages. Peripheral artery disease is a globally prevalent cardiovascular disease affecting millions of people. Progression of the disease leads to intermittent claudication, subsequent critical limb ischemia and muscle injury. Tissue-derived and ex vivo-expanded mesenchymal stromal cells (MSCs) for skeletal muscle regeneration have been studied, but pre-clinical and clinical results have not been consistent. As a result, the potential therapeutic efficacy and associated repair mechanisms of MSCs remain unclear. Numerous studies have demonstrated the vulnerability of delivered MSCs, with a precipitous drop in cell viability upon transplantation. This has prompted investigation into the therapeutic benefit of apoptotic cells, microvesicles, exosomes and soluble signals that are released upon cell death. METHODS In this study, we characterized various components produced by MSCs after cell death induction under different conditions. We discovered anti-inflammatory and pro-regenerative effects produced by cell components following a freeze and thaw (F&T) process on macrophage polarization in vitro. We further investigated the underlying mechanisms of macrophage polarization by those components resulting from severe cell death induction. RESULTS We found potent therapeutic effects from F&T-induced cell debris are dependent on the externalization of phosphatidylserine on the plasma membrane. In contrast, effects from the supernatant of F&T-induced cell death primarily depends on the released protein content. We then applied the F&T-induced cell supernatant to an animal model of peripheral artery disease to treat muscle injury caused by severe ischemia. Treatment with the F&T supernatant but not the vulnerable MSCs resulted in significantly improved recovery of muscle function, blood flow and morphology and inflammation resolution in the affected muscles 2 weeks after injury. CONCLUSIONS This study validates the therapeutic potential of F&T-induced supernatant obviating the need for a viable population from vulnerable MSCs to treat injury, thus providing a roadmap for cell-free therapeutic approaches for tissue regeneration.
Collapse
Affiliation(s)
- Wenbai Huang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA; Department of Kinesiology and Health Education, University of Texas at Austin, Austin, Texas, USA
| | - Chelsea A Kraynak
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Elizabeth C Bender
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Roger P Farrar
- Department of Kinesiology and Health Education, University of Texas at Austin, Austin, Texas, USA
| | - Laura J Suggs
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas, USA.
| |
Collapse
|
8
|
McPherson CA, Kelly-Rajan K, Lefebvre d’Hellencourt C, Harry GJ. High Content Imaging and Quantification of Microglia Phagocytosis In Vitro. Curr Protoc 2023; 3:e638. [PMID: 36622815 PMCID: PMC10035415 DOI: 10.1002/cpz1.638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Microglia function as the tissue-specific resident macrophages of the nervous system, performing immune and non-immune functions. These functions are critical to development and to maintain homeostasis in the nervous system throughout the lifespan, and during brain injury or disease. One method by which microglia maintain homeostasis is phagocytosis of aberrant proteins, extracellular debris, synapses, or apoptotic cells. Phagocytic function can be changed by environmental or genetic risk factors that affect microglia. These protocols present a rapid and simple in vitro high-content imaging protocol for studying phagocytosis in the murine microglia BV-2 cell line. High-content imaging and analysis enable versatility of the assay, which can be used to test multiple experimental conditions, or as a screening tool. © 2023 Wiley Periodicals LLC. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA. Basic Protocol 1: Phagocytosis of fluorescently labeled particles Basic Protocol 2: Examining modifications to phagocytosis by test substances Basic Protocol 3: High content imaging and analysis of phagocytic cells.
Collapse
Affiliation(s)
- Christopher A. McPherson
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Kiran Kelly-Rajan
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
- University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christian Lefebvre d’Hellencourt
- Inserm, UMR 1188 Diabète Athérothrombose Thérapies Réunion Océan Indien (DéTROI), plateforme CYROI, 97490 Sainte-Clotilde, France; Université de La Réunion, UMR 1188, 97490 Sainte-Clotilde, France
| | - G. Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| |
Collapse
|
9
|
Loh W, Vermeren S. Anti-Inflammatory Neutrophil Functions in the Resolution of Inflammation and Tissue Repair. Cells 2022; 11:cells11244076. [PMID: 36552840 PMCID: PMC9776979 DOI: 10.3390/cells11244076] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Neutrophils are highly abundant circulating leukocytes that are amongst the first cells to be recruited to sites of infection or sterile injury. Their ability to generate and release powerful cytotoxic products ties with their role in host defence from bacterial and fungal infections. Neutrophilic inflammation is tightly regulated to limit the amount of 'bystander injury' caused. Neutrophils were in the past regarded as short-lived, indiscriminate killers of invading microorganisms. However, this view has changed quite dramatically in recent years. Amongst other insights, neutrophils are now recognised to also have important anti-inflammatory functions that are critical for the resolution of inflammation and return to homeostasis. This minireview focusses on anti-inflammatory neutrophil functions, placing a particular focus on recent findings linked to neutrophil cell death, several types of which may be anti-inflammatory (apoptosis, secondary necrosis, and neutrophil extracellular traps). These are discussed together with features that may further promote the clearance of dead cells by efferocytosis and reprogramming of macrophages to promote resolution and repair.
Collapse
Affiliation(s)
- Waywen Loh
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH10 5HF, UK
| | - Sonja Vermeren
- Centre for Inflammation Research, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh EH10 5HF, UK
| |
Collapse
|
10
|
Eating the Enemy: Mycoplasma Strategies to Evade Neutrophil Extracellular Traps (NETs) Promoting Bacterial Nucleotides Uptake and Inflammatory Damage. Int J Mol Sci 2022; 23:ijms232315030. [PMID: 36499356 PMCID: PMC9740415 DOI: 10.3390/ijms232315030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Neutrophils are effector cells involved in the innate immune response against infection; they kill infectious agents in the intracellular compartment (phagocytosis) or in the extracellular milieu (degranulation). Moreover, neutrophils release neutrophil extracellular traps (NETs), complex structures composed of a scaffold of decondensed DNA associated with histones and antimicrobial compounds; NETs entrap infectious agents, preventing their spread and promoting their clearance. NET formation is triggered by microbial compounds, but many microorganisms have evolved several strategies for NET evasion. In addition, the dysregulated production of NETs is associated with chronic inflammatory diseases. Mycoplasmas are reduced genome bacteria, able to induce chronic infections with recurrent inflammatory symptoms. Mycoplasmas' parasitic lifestyle relies on metabolite uptake from the host. Mycoplasmas induce NET release, but their surface or secreted nucleases digest the NETs' DNA scaffold, allowing them to escape from entrapment and providing essential nucleotide precursors, thus promoting the infection. The presence of Mycoplasma species has been associated with chronic inflammatory disorders, such as systemic lupus erythematosus, rheumatoid arthritis, inflammatory bowel disease, Crohn's disease, and cancer. The persistence of mycoplasma infection and prolonged NET release may contribute to the onset of chronic inflammatory diseases and needs further investigation and insights.
Collapse
|
11
|
Sun JL, Lyu TB, Chen ZL, Lian CF, Liu SY, Shao TH, Zhang S, Zhao LL, Liu JJ, Peng LY, Zhang L, Yang YJ, Zhang FC, Chen H. Methylprednisolone pulse therapy promotes the differentiation of regulatory T cells by inducing the apoptosis of CD4 + T cells in patients with systemic lupus erythematosus. Clin Immunol 2022; 241:109079. [PMID: 35842211 DOI: 10.1016/j.clim.2022.109079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 06/28/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES To investigate the differentiation of regulatory T cells (Tregs) induced by methylprednisolone (MP) pulse therapy in patients with Systemic Lupus Erythematosus (SLE). METHODS We enrolled 30 patients with SLE and analyzed peripheral blood mononuclear cells (PBMCs) before and after MP pulse therapy. Peripheral Tregs, apoptosis of PBMCs subsets, and TGFβ production by monocytes was quantified by flow cytometry. Proliferation and IFN-γ production of CD4+ T cells were measured. Furthermore, TGFβ1 production by human monocyte-derived macrophages (HMDM) stimulated with MP-treated CD4+ T cells were quantified by ELISA. RESULTS Peripheral Tregs was significantly increased after MP pulse therapy (6.76 ± 1.46% vs. 3.82 ± 1.02%, p < 0.01), with an expansion of Nrp1- induced Tregs (4.54 ± 0.46% vs. 1.75 ± 0.38%, p < 0.01). Proliferation and IFN-γ production of CD4+ T cells were significantly decreased after MP pulse therapy. MP pulse therapy induced CD4+ T cell apoptosis (early apoptosis, 26.34 ± 3.54% vs. 14.81 ± 2.89%, p < 0.01) and TGFβ expression on monocytes (6.02% vs. 2.45%, p < 0.01). Furthermore, MP induced CD4+ T cell apoptosis in vitro, which stimulated HMDM to produce TGFβ. Moreover, elevated TGFβ level in supernatant from HMDM stimulated with MP-treated CD4+ T cells promoted Tregs differentiation. CONCLUSIONS MP pulse therapy induces CD4+ T cell apoptosis, which promotes monocytes to produce TGFβ and further facilitates Tregs differentiation. Newly-differentiated Tregs suppress proliferation and IFN-γ production of CD4+ T cells and contribute to immunoregulatory milieu after MP pulse therapy.
Collapse
Affiliation(s)
- Jin-Lei Sun
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China; Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tai-Biao Lyu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China
| | - Zhi-Lei Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China
| | - Chao-Feng Lian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China; Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Su-Ying Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China
| | - Ti-Hong Shao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China
| | - Shuo Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China
| | - Li-Ling Zhao
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China
| | - Jin-Jing Liu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China; National Clinical Center for Dermatologic and Autoimmune Disorders, China
| | - Lin-Yi Peng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China; National Clinical Center for Dermatologic and Autoimmune Disorders, China
| | - Li Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China; National Clinical Center for Dermatologic and Autoimmune Disorders, China
| | - Yun-Jiao Yang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China; National Clinical Center for Dermatologic and Autoimmune Disorders, China
| | - Feng-Chun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China; National Clinical Center for Dermatologic and Autoimmune Disorders, China
| | - Hua Chen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, China; National Clinical Center for Dermatologic and Autoimmune Disorders, China.
| |
Collapse
|
12
|
Lee YJ, Kim K, Kim M, Ahn YH, Kang JL. Inhibition of STAT6 Activation by AS1517499 Inhibits Expression and Activity of PPARγ in Macrophages to Resolve Acute Inflammation in Mice. Biomolecules 2022; 12:447. [PMID: 35327639 PMCID: PMC8946515 DOI: 10.3390/biom12030447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/07/2022] [Accepted: 03/12/2022] [Indexed: 12/10/2022] Open
Abstract
Signal transducer and activator of transcription 6 (STAT6) promotes an anti-inflammatory process by inducing the development of M2 macrophages. We investigated whether modulating STAT6 activity in macrophages using AS1517499, the specific STAT6 inhibitor, affects the restoration of homeostasis after an inflammatory insult by regulating PPARγ expression and activity. Administration of AS1517499 suppressed the enhanced STAT6 phosphorylation and nuclear translocation observed in peritoneal macrophages after zymosan injection. In addition, AS1517499 delayed resolution of acute inflammation as evidenced by enhanced secretion of pro-inflammatory cytokines, reduced secretion of anti-inflammatory cytokines in PLF and supernatants from peritoneal macrophages, and exaggerated neutrophil numbers and total protein levels in PLF. We demonstrate temporal increases in annexin A1 (AnxA1) protein and mRNA levels in peritoneal lavage fluid (PLF), peritoneal macrophages, and spleen in a murine model of zymosan-induced acute peritonitis. In vitro priming of mouse bone marrow-derived macrophages (BMDM) and peritoneal macrophages with AnxA1 induced STAT6 activation with enhanced PPARγ expression and activity. Using AS1517499, we demonstrate that inhibition of STAT6 activation delayed recovery of PPARγ expression and activity, as well as impaired efferocytosis. Taken together, these results suggest that activation of the STAT6 signaling pathway mediates PPARγ expression and activation in macrophages to resolve acute inflammation.
Collapse
Affiliation(s)
- Ye-Ji Lee
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (Y.-J.L.); (K.K.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (M.K.); (Y.-H.A.)
| | - Kiyoon Kim
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (Y.-J.L.); (K.K.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (M.K.); (Y.-H.A.)
| | - Minsuk Kim
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (M.K.); (Y.-H.A.)
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Young-Ho Ahn
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (M.K.); (Y.-H.A.)
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Jihee Lee Kang
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (Y.-J.L.); (K.K.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (M.K.); (Y.-H.A.)
| |
Collapse
|
13
|
Marković D, Maslovarić I, Djikić D, Čokić VP. Neutrophil Death in Myeloproliferative Neoplasms: Shedding More Light on Neutrophils as a Pathogenic Link to Chronic Inflammation. Int J Mol Sci 2022; 23:1490. [PMID: 35163413 PMCID: PMC8836089 DOI: 10.3390/ijms23031490] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 12/15/2022] Open
Abstract
Neutrophils are an essential component of the innate immune response, but their prolonged activation can lead to chronic inflammation. Consequently, neutrophil homeostasis is tightly regulated through balance between granulopoiesis and clearance of dying cells. The bone marrow is both a site of neutrophil production and the place they return to and die. Myeloproliferative neoplasms (MPN) are clonal hematopoietic disorders characterized by the mutations in three types of molecular markers, with emphasis on Janus kinase 2 gene mutation (JAK2V617F). The MPN bone marrow stem cell niche is a site of chronic inflammation, with commonly increased cells of myeloid lineage, including neutrophils. The MPN neutrophils are characterized by the upregulation of JAK target genes. Additionally, MPN neutrophils display malignant nature, they are in a state of activation, and with deregulated apoptotic machinery. In other words, neutrophils deserve to be placed in the midst of major events in MPN. Our crucial interest in this review is better understanding of how neutrophils die in MPN mirrored by defects in apoptosis and to what possible extent they can contribute to MPN pathophysiology. We tend to expect that reduced neutrophil apoptosis will establish a pathogenic link to chronic inflammation in MPN.
Collapse
Affiliation(s)
- Dragana Marković
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, POB 39, 11129 Belgrade, Serbia;
| | - Irina Maslovarić
- Group for Immunology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, POB 39, 11129 Belgrade, Serbia;
| | - Dragoslava Djikić
- Group for Molecular Oncology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, POB 39, 11129 Belgrade, Serbia; (D.D.); (V.P.Č.)
| | - Vladan P. Čokić
- Group for Molecular Oncology, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr Subotića 4, POB 39, 11129 Belgrade, Serbia; (D.D.); (V.P.Č.)
| |
Collapse
|
14
|
Morana O, Wood W, Gregory CD. The Apoptosis Paradox in Cancer. Int J Mol Sci 2022; 23:ijms23031328. [PMID: 35163253 PMCID: PMC8836235 DOI: 10.3390/ijms23031328] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer growth represents a dysregulated imbalance between cell gain and cell loss, where the rate of proliferating mutant tumour cells exceeds the rate of those that die. Apoptosis, the most renowned form of programmed cell death, operates as a key physiological mechanism that limits cell population expansion, either to maintain tissue homeostasis or to remove potentially harmful cells, such as those that have sustained DNA damage. Paradoxically, high-grade cancers are generally associated with high constitutive levels of apoptosis. In cancer, cell-autonomous apoptosis constitutes a common tumour suppressor mechanism, a property which is exploited in cancer therapy. By contrast, limited apoptosis in the tumour-cell population also has the potential to promote cell survival and resistance to therapy by conditioning the tumour microenvironment (TME)-including phagocytes and viable tumour cells-and engendering pro-oncogenic effects. Notably, the constitutive apoptosis-mediated activation of cells of the innate immune system can help orchestrate a pro-oncogenic TME and may also effect evasion of cancer treatment. Here, we present an overview of the implications of cell death programmes in tumour biology, with particular focus on apoptosis as a process with "double-edged" consequences: on the one hand, being tumour suppressive through deletion of malignant or pre-malignant cells, while, on the other, being tumour progressive through stimulation of reparatory and regenerative responses in the TME.
Collapse
|
15
|
Asare PF, Tran HB, Hurtado PR, Perkins GB, Nguyen P, Jersmann H, Roscioli E, Hodge S. Inhibition of LC3-associated phagocytosis in COPD and in response to cigarette smoke. Ther Adv Respir Dis 2021; 15:17534666211039769. [PMID: 34852704 PMCID: PMC8647217 DOI: 10.1177/17534666211039769] [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] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION/RATIONALE In chronic obstructive pulmonary disease (COPD), defective macrophage phagocytic clearance of cells undergoing apoptosis by efferocytosis may lead to secondary necrosis of the uncleared cells and contribute to airway inflammation. The precise mechanisms for this phenomenon remain unknown. LC3-associated phagocytosis (LAP) is indispensable for effective efferocytosis. We hypothesized that cigarette smoke inhibits the regulators of LAP pathway, potentially contributing to the chronic airways inflammation associated with COPD. METHODS Bronchoalveolar (BAL)-derived alveolar macrophages, lung tissue macrophages obtained from lung resection surgery, and monocyte-derived macrophages (MDM) were prepared from COPD patients and control participants. Lung/airway samples from mice chronically exposed to cigarette smoke were also investigated. Differentiated THP-1 cells were exposed to cigarette smoke extract (CSE). The LAP pathway including Rubicon, as an essential regulator of LAP, efferocytosis and inflammation was examined using western blot, ELISA, flow cytometry, and/or immunofluorescence. RESULTS Rubicon was significantly depleted in COPD alveolar macrophages compared with non-COPD control macrophages. Rubicon protein in alveolar macrophages of cigarette smoke-exposed mice and cigarette smoke-exposed MDM and THP-1 was decreased with a concomitant impairment of efferocytosis. We also noted increased expression of LC3 which is critical for LAP pathway in COPD and THP-1 macrophages. Furthermore, THP-1 macrophages exposed to cigarette smoke extract exhibited higher levels of other key components of LAP pathway including Atg5 and TIM-4. There was a strong positive correlation between Rubicon protein expression and efferocytosis. CONCLUSION LAP is a requisite for effective efferocytosis and an appropriate inflammatory response, which is impaired by Rubicon deficiency. Our findings suggest dysregulated LAP due to reduced Rubicon as a result of CSE exposure. This phenomenon could lead to a failure of macrophages to effectively process phagosomes containing apoptotic cells during efferocytosis. Restoring Rubicon protein expression has unrecognized therapeutic potential in the context of disease-related modifications caused by exposure to cigarette smoke.
Collapse
Affiliation(s)
- Patrick F Asare
- Department of Thoracic Medicine, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Hai B Tran
- Department of Thoracic Medicine, School of Medicine, The University of Adelaide, Adelaide, SA, Australia
| | - Plinio R Hurtado
- Department of Renal Medicine, School of Medicine, The University of Adelaide, Adelaide, SA, Australia.,Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Griffith B Perkins
- Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Phan Nguyen
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Hubertus Jersmann
- Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Eugene Roscioli
- Department of Thoracic Medicine, School of Medicine, The University of Adelaide, Adelaide, SA, Australia.,Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Sandra Hodge
- Department of Thoracic Medicine, Faculty of Health and Medical Science, The University of Adelaide, Adelaide, SA 5005, Australia.,School of Medicine, The University of Adelaide, Adelaide, SA, Australia.,Department of Thoracic Medicine, Royal Adelaide Hospital, Adelaide, SA, Australia
| |
Collapse
|
16
|
Juban G, Chazaud B. Efferocytosis during Skeletal Muscle Regeneration. Cells 2021; 10:cells10123267. [PMID: 34943775 PMCID: PMC8699096 DOI: 10.3390/cells10123267] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 02/07/2023] Open
Abstract
Efferocytosis, i.e., engulfment of dead cells by macrophages, is a crucial step during tissue repair after an injury. Efferocytosis delineates the transition from the pro-inflammatory phase of the inflammatory response to the recovery phase that ensures tissue reconstruction. We present here the role of efferocytosis during skeletal muscle regeneration, which is a paradigm of sterile tissue injury followed by a complete regeneration. We present the molecular mechanisms that have been described to control this process, and particularly the metabolic control of efferocytosis during skeletal muscle regeneration.
Collapse
|
17
|
Singhal A, Kumar S. Neutrophil and remnant clearance in immunity and inflammation. Immunology 2021; 165:22-43. [PMID: 34704249 DOI: 10.1111/imm.13423] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/18/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022] Open
Abstract
Neutrophil-centred inflammation and flawed clearance of neutrophils cause and exuberate multiple pathological conditions. These most abundant leukocytes exhibit very high daily turnover in steady-state and stress conditions. Various armours including oxidative burst, NETs and proteases function against pathogens, but also dispose neutrophils to spawn pro-inflammatory responses. Neutrophils undergo death through different pathways upon ageing, infection, executing the intruder's elimination. These include non-lytic apoptosis and other lytic deaths including NETosis, necroptosis and pyroptosis with distinct disintegration of the cellular membrane. This causes release and presence of different intracellular cytotoxic, and tissue-damaging content as cell remnants in the extracellular environment. The apoptotic cells and apoptotic bodies get cleared with non-inflammatory outcomes, while lytic deaths associated remnants including histones and cell-free DNA cause pro-inflammatory responses. Indeed, the enhanced frequencies of neutrophil-associated proteases, cell-free DNA and autoantibodies in diverse pathologies including sepsis, asthma, lupus and rheumatoid arthritis, imply disturbed neutrophil resolution programmes in inflammatory and autoimmune diseases. Thus, the clearance mechanisms of neutrophils and associated remnants are vital for therapeutics. Though studies focused on clearance mechanisms of senescent or apoptotic neutrophils so far generated a good understanding of the same, clearance of neutrophils undergoing distinct lytic deaths, including NETs, are being the subjects of intense investigations. Here, in this review, we are providing the current updates in the clearance mechanisms of apoptotic neutrophils and focusing on not so well-defined recognition, uptake and degradation of neutrophils undergoing lytic death and associated remnants that may provide new therapeutic approaches in inflammation and autoimmunity.
Collapse
Affiliation(s)
- Apurwa Singhal
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Sachin Kumar
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India.,Academy of Scientific and Innovative Research (AcSIR), Postal Staff College Area, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
18
|
Monteith AJ, Miller JM, Maxwell CN, Chazin WJ, Skaar EP. Neutrophil extracellular traps enhance macrophage killing of bacterial pathogens. SCIENCE ADVANCES 2021; 7:eabj2101. [PMID: 34516771 PMCID: PMC8442908 DOI: 10.1126/sciadv.abj2101] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/20/2021] [Indexed: 05/16/2023]
Abstract
Neutrophils and macrophages are critical to the innate immune response, but cooperative mechanisms used by these cells to combat extracellular pathogens are not well understood. This study reveals that S100A9-deficient neutrophils produce higher levels of mitochondrial superoxide in response to Staphylococcus aureus and, as a result, form neutrophil extracellular traps (suicidal NETosis). Increased suicidal NETosis does not improve neutrophil killing of S. aureus in isolation but augments macrophage killing. NET formation enhances antibacterial activity by increasing phagocytosis by macrophages and by transferring neutrophil-specific antimicrobial peptides to them. Similar results were observed in response to other phylogenetically distinct bacterial pathogens including Streptococcus pneumoniae and Pseudomonas aeruginosa, implicating this as an immune defense mechanism that broadly enhances antibacterial activity. These results demonstrate that achieving maximal bactericidal activity through NET formation requires macrophages and that accelerated and more robust suicidal NETosis makes neutrophils adept at increasing antibacterial activity, especially when A9 deficient.
Collapse
Affiliation(s)
- Andrew J. Monteith
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeanette M. Miller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - C. Noel Maxwell
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
| | - Walter J. Chazin
- Department of Biochemistry, Vanderbilt University, Nashville, TN, USA
- Center for Structural Biology, Vanderbilt University, Nashville, TN, USA
- Department of Chemistry, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P. Skaar
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
19
|
Luk HY, Levitt DE, Appell C, Vingren JL. Sex Dimorphism in Muscle Damage-induced Inflammation. Med Sci Sports Exerc 2021; 53:1595-1605. [PMID: 34261990 DOI: 10.1249/mss.0000000000002628] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The purpose of this study was to determine the effect of resistance exercise (RE)-induced hormonal changes on intramuscular cytokine gene expression after muscle damage in untrained men and women. METHODS Men (n = 8, 22 ± 3 yr) and women (n = 8, 19 ± 1 yr) completed two sessions of 80 unilateral maximal eccentric knee extensions followed by either an upper body RE bout (EX) or a time-matched period (CON). Muscle samples (vastus laterals) were analyzed for mRNA expression of interleukin (IL) 6, IL-10, IL-15, TNFA, TGFB, CCL2, and CD68 at PRE, 12 h, and 24 h after the session. RESULTS A significant time-sex-condition interaction was found for TGFB with an increase for EX in men at 12 h from PRE. For EX, TGFB was also greater in men than in women at 12 and 24 h. Significant time-sex and condition-sex interactions were found for IL-10 with an increase for men that was greater than for women at 12 and 24 h. IL-10 was lower in EX than CON for men. A significant time-sex interaction was found for TNFA with an increase for men that was greater than for women at 24 h. A significant time-condition interaction was found for CD68 with an increase at 12 h and decrease at 24 h for EX and CON. CD68 was lower in EX than CON at 12 h. A significant time effect was found for IL6 and CCL2 with an increase at 12 and 24 h. CONCLUSIONS Results suggest that women seem to have a muted intramuscular cytokine (i.e., IL-10, TNF-α, and TGF-β) response to muscle damage compared with men.
Collapse
Affiliation(s)
- Hui-Ying Luk
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX
| | | | - Casey Appell
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX
| | - Jakob L Vingren
- Applied Physiology Laboratory, Department of Kinesiology, Health Promotion, and Recreation, University of North Texas, Denton, TX
| |
Collapse
|
20
|
Abstract
Bronchiectasis is a complex, heterogeneous disorder defined by both a radiological abnormality of permanent bronchial dilatation and a clinical syndrome. There are multiple underlying causes including severe infections, mycobacterial disease, autoimmune conditions, hypersensitivity disorders, and genetic conditions. The pathophysiology of disease is understood in terms of interdependent concepts of chronic infection, inflammation, impaired mucociliary clearance, and structural lung damage. Neutrophilic inflammation is characteristic of the disease, with elevated levels of harmful proteases such as neutrophil elastase associated with worse outcomes. Recent data show that neutrophil extracellular trap formation may be the key mechanism leading to protease release and severe bronchiectasis. Despite the dominant of neutrophilic disease, eosinophilic subtypes are recognized and may require specific treatments. Neutrophilic inflammation is associated with elevated bacterial loads and chronic infection with organisms such as Pseudomonas aeruginosa. Loss of diversity of the normal lung microbiota and dominance of proteobacteria such as Pseudomonas and Haemophilus are features of severe bronchiectasis and link to poor outcomes. Ciliary dysfunction is also a key feature, exemplified by the rare genetic syndrome of primary ciliary dyskinesia. Mucus symptoms arise through goblet cell hyperplasia and metaplasia and reduced ciliary function through dyskinesia and loss of ciliated cells. The contribution of chronic inflammation, infection, and mucus obstruction leads to progressive structural lung damage. The heterogeneity of the disease is the most challenging aspect of management. An understanding of the pathophysiology of disease and their biomarkers can help to guide personalized medicine approaches utilizing the concept of "treatable traits."
Collapse
Affiliation(s)
- Holly R Keir
- Scottish Centre for Respiratory Research, University of Dundee, Dundee, United Kingdom
| | - James D Chalmers
- Scottish Centre for Respiratory Research, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
21
|
Bassin EJ, Piganelli JD, Little SR. Auto-antigen and Immunomodulatory Agent-Based Approaches for Antigen-Specific Tolerance in NOD Mice. Curr Diab Rep 2021; 21:9. [PMID: 33547977 DOI: 10.1007/s11892-021-01376-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 10/22/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) can be managed by insulin replacement, but it is still associated with an increased risk of microvascular/cardiovascular complications. There is considerable interest in antigen-specific approaches for treating T1D due to their potential for a favorable risk-benefit ratio relative to non-specific immune-based treatments. Here we review recent antigen-specific tolerance approaches using auto-antigen and/or immunomodulatory agents in NOD mice and provide insight into seemingly contradictory findings. RECENT FINDINGS Although delivery of auto-antigen alone can prevent T1D in NOD mice, this approach may be prone to inconsistent results and has not demonstrated an ability to reverse established T1D. Conversely, several approaches that promote presentation of auto-antigen in a tolerogenic context through cell/tissue targeting, delivery system properties, or the delivery of immunomodulatory agents have had success in reversing recent-onset T1D in NOD mice. While initial auto-antigen based approaches were unable to substantially influence T1D progression clinically, recent antigen-specific approaches have promising potential.
Collapse
Affiliation(s)
- Ethan J Bassin
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jon D Piganelli
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh, 4401 Penn Avenue, 6125 Rangos Research Center, Pittsburgh, PA, 15224, USA.
| | - Steven R Little
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Chemical Engineering, University of Pittsburgh, 3700 O'Hara Street, 940 Benedum Hall, Pittsburgh, PA, 15261, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.
- Department of Pharmaceutical Science, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
Zhou Y, Wang Y, Chen H, Xu Y, Luo Y, Deng Y, Zhang J, Shao A. Immuno-oncology: are TAM receptors in glioblastoma friends or foes? Cell Commun Signal 2021; 19:11. [PMID: 33509214 PMCID: PMC7841914 DOI: 10.1186/s12964-020-00694-8] [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: 09/22/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022] Open
Abstract
Tyro3, Axl, and Mertk (TAM) receptors are a subfamily of receptor tyrosine kinases. TAM receptors have been implicated in mediating efferocytosis, regulation of immune cells, secretion of inflammatory factors, and epithelial-to-mesenchymal transition in the tumor microenvironment, thereby serving as a critical player in tumor development and progression. The pro-carcinogenic role of TAM receptors has been widely confirmed, overexpression of TAM receptors is tied to tumor cells growth, metastasis, invasion and treatment resistance. Nonetheless, it is surprising to detect that inhibiting TAM signaling is not all beneficial in the tumor immune microenvironment. The absence of TAM receptors also affects anti-tumor immunity under certain conditions by modulating different immune cells, as the functional diversification of TAM signaling is closely related to tumor immunotherapy. Glioblastoma is the most prevalent and lethal primary brain tumor in adults. Although research regarding the crosstalk between TAM receptors and glioblastoma remains scarce, it appears likely that TAM receptors possess potential anti-tumor effects rather than portraying a total cancer-driving role in the context of glioblastoma. Accordingly, we doubt whether TAM receptors play a double-sided role in glioblastoma, and propose the Janus-faced TAM Hypothesis as a conceptual framework for comprehending the precise underlying mechanisms of TAMs. In this study, we aim to cast a spotlight on the potential multidirectional effects of TAM receptors in glioblastoma and provide a better understanding for TAM receptor-related targeted intervention. Video Abstract
Collapse
Affiliation(s)
- Yunxiang Zhou
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yali Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hailong Chen
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Yanyan Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211126, Jiangsu, China
| | - Yi Luo
- The Second Affiliated Hospital of Zhejiang University School of Medicine (Changxing Branch), Changxing, Huzhou, 313100, Zhejiang, China
| | - Yongchuan Deng
- Department of Surgical Oncology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
| |
Collapse
|
23
|
Tajbakhsh A, Farahani N, Gheibihayat SM, Mirkhabbaz AM, Savardashtaki A, Hamblin MR, Mirzaei H. Autoantigen-specific immune tolerance in pathological and physiological cell death: Nanotechnology comes into view. Int Immunopharmacol 2020; 90:107177. [PMID: 33249046 DOI: 10.1016/j.intimp.2020.107177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023]
Abstract
Apoptotic cells are tolerogenic and can present self-antigens in the absence of inflammation, to antigen-presenting cells by the process of efferocytosis, resulting in anergy and depletion of immune effector cells. This tolerance is essential to maintain immune homeostasis and prevent systemic autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus. Consequently, effective efferocytosis can result in the induction of immune tolerance mediated via triggering modulatory lymphocytes and anti-inflammatory responses. Furthermore, several distinct soluble factors, receptors and pathways have been found to be involved in the efferocytosis, which are able to regulate immune tolerance by lessening antigen presentation, inhibition of T-cell proliferation and induction of regulatory T-cells. Some newly developed nanotechnology-based approaches can induce antigen-specific immunological tolerance without any systemic immunosuppression. These strategies have been explored to reverse autoimmune responses induced against various protein antigens in different diseases. In this review, we describe some nanotechnology-based approaches for the maintenance of self-tolerance using the apoptotic cell clearance process (efferocytosis) that may be able to induce immune tolerance and treat autoimmune diseases.
Collapse
Affiliation(s)
- Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Farahani
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sayed Mohammad Gheibihayat
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, Johannesburg 2028, South Africa.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R., Iran.
| |
Collapse
|
24
|
Lee J, Lu Y, Oshins R, West J, Moneypenny CG, Han K, Brantly ML. Alpha 1 Antitrypsin-Deficient Macrophages Have Impaired Efferocytosis of Apoptotic Neutrophils. Front Immunol 2020; 11:574410. [PMID: 33329539 PMCID: PMC7714766 DOI: 10.3389/fimmu.2020.574410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/27/2020] [Indexed: 12/05/2022] Open
Abstract
Alpha 1 antitrypsin deficiency (AATD) is an autosomal co-dominant disorder characterized by a low level of circulating AAT, which significantly reduces protection for the lower airways against proteolytic burden caused by neutrophils. Neutrophils, which are terminally differentiated innate immune cells and play a critical role to clear pathogens, accumulate excessively in the lung of AATD individuals. The neutrophil burden in AATD individuals increases the risk for early-onset destructive lung diseases by producing neutrophil products such as reactive oxygen radicals and various proteases. The level of AAT in AATD individuals is not sufficient to inhibit the activity of neutrophil chemotactic factors such as CXCL-8 and LTB4, which could lead to alveolar neutrophil accumulation in AATD individuals. However, as neutrophils have a short lifespan, and apoptotic neutrophils are rapidly cleared by alveolar macrophages that outnumber the apoptotic neutrophils in the pulmonary alveolus, the increased chemotaxis activity does not fully explain the persistent neutrophil accumulation and the resulting chronic inflammation in AATD individuals. Here, we propose that the ability of alveolar macrophages to clear apoptotic neutrophils is impaired in AATD individuals and it could be the main driver to cause neutrophil accumulation in their lung. This study demonstrates that Z-AAT variant significantly increases the expression of pro-inflammatory cytokines including CXCL-8, CXCL1, LTB4, and TNFα in LPS-treated macrophages. These cytokines play a central role in neutrophil recruitment to the lung and in clearance of apoptotic neutrophils by macrophages. Our result shows that LPS treatment significantly reduces the efferocytosis ability of macrophages with the Z-AAT allele by inducing TNFα expression. We incubated monocyte-derived macrophages (MDMs) with apoptotic neutrophils and found that after 3 h of co-incubation, the expression level of CXCL-8 is reduced in M-MDMs but increased in Z-MDMs. This result shows that the expression of inflammatory cytokines could be increased by impaired efferocytosis. It indicates that the efferocytosis ability of macrophages plays an important role in regulating cytokine expression and resolving inflammation. Findings from this study would help us better understand the multifaceted effect of AAT on regulating neutrophil balance in the lung and the underlying mechanisms.
Collapse
Affiliation(s)
- Jungnam Lee
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Yuanqing Lu
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Regina Oshins
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Jesse West
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Craig G Moneypenny
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| | - Kyudong Han
- Department of Microbiology, College of Science and Technology, Dankook University, Cheonan, South Korea.,DKU-Theragen Institute for NGS Analysis, Cheonan, South Korea
| | - Mark L Brantly
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
25
|
Arnhold J. The Dual Role of Myeloperoxidase in Immune Response. Int J Mol Sci 2020; 21:E8057. [PMID: 33137905 PMCID: PMC7663354 DOI: 10.3390/ijms21218057] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/25/2020] [Accepted: 10/28/2020] [Indexed: 12/14/2022] Open
Abstract
The heme protein myeloperoxidase (MPO) is a major constituent of neutrophils. As a key mediator of the innate immune system, neutrophils are rapidly recruited to inflammatory sites, where they recognize, phagocytose, and inactivate foreign microorganisms. In the newly formed phagosomes, MPO is involved in the creation and maintenance of an alkaline milieu, which is optimal in combatting microbes. Myeloperoxidase is also a key component in neutrophil extracellular traps. These helpful properties are contrasted by the release of MPO and other neutrophil constituents from necrotic cells or as a result of frustrated phagocytosis. Although MPO is inactivated by the plasma protein ceruloplasmin, it can interact with negatively charged components of serum and the extracellular matrix. In cardiovascular diseases and many other disease scenarios, active MPO and MPO-modified targets are present in atherosclerotic lesions and other disease-specific locations. This implies an involvement of neutrophils, MPO, and other neutrophil products in pathogenesis mechanisms. This review critically reflects on the beneficial and harmful functions of MPO against the background of immune response.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Institute of Medical Physics and Biophysics, Medical Faculty, Leipzig University, 04 107 Leipzig, Germany
| |
Collapse
|
26
|
Neutrophil elastase promotes macrophage cell adhesion and cytokine production through the integrin-Src kinases pathway. Sci Rep 2020; 10:15874. [PMID: 32981934 PMCID: PMC7522083 DOI: 10.1038/s41598-020-72667-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/26/2020] [Indexed: 01/08/2023] Open
Abstract
There are a number of respiratory diseases characterized by the presence of excess neutrophil elastase (NE) activity in tissues, including cystic fibrosis and chronic obstructive pulmonary disease (COPD). NE is considered a primary contributor to disease development, but the precise mechanism has yet to be fully determined. We hypothesized that NE alters the function of macrophages (Mɸ) which play a critical role in many physiological processes in healthy lungs. We demonstrate that monocyte-derived Mɸ exposed to NE releases active matrix metalloproteinases (MMPs), increase expression of pro-inflammatory cytokines TNFα, IL-1β, and IL-8, and reduce capacity to phagocytose bacteria. Changes in Mɸ function following NE treatment were accompanied by increased adhesion and cytoskeleton re-arrangement, indicating the possibility of integrin involvement. To support this observation, we demonstrate that NE induces phosphorylation of kinases from the Src kinase family, a hallmark of integrin signaling activation. Moreover, pretreatment of Mɸ with a specific Src kinase inhibitor, PP2 completely prevents NE-induced pro-inflammatory cytokine production. Taken together these findings indicate that NE participates in lung destruction not only through direct proteolytic degradation of matrix proteins, but also through activation of Mɸ inflammatory and proteolytic functions.
Collapse
|
27
|
Cornish SM, Chilibeck PD, Candow DG. Potential Importance of Immune System Response to Exercise on Aging Muscle and Bone. Curr Osteoporos Rep 2020; 18:350-356. [PMID: 32500480 DOI: 10.1007/s11914-020-00596-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The age-related loss of skeletal muscle and bone tissue decreases functionality and increases the risk for falls and injuries. One contributing factor of muscle and bone loss over time is chronic low-grade inflammation. Exercise training is an effective countermeasure for decreasing the loss of muscle and bone tissue, possibly by enhancing immune system response. Herein, we discuss key interactions between the immune system, muscle, and bone in relation to exercise perturbations, and we identify that there is substantial "cross-talk" between muscle and bone and the immune system in response to exercise. RECENT FINDINGS Recent advances in our understanding of the "cross-talk" between muscle and bone and the immune system indicate that exercise is likely to mediate many of the beneficial effects on muscle and bone via their interactions with the immune system. The age-related loss of muscle and bone tissue may be partially explained by an impaired immune system via chronic low-grade inflammation. Exercise training has a beneficial effect on immune system function and aging muscle and bone. Theoretically, the "cross-talk" between the immune system, muscle, and bone in response to exercise enhances aging musculoskeletal health.
Collapse
Affiliation(s)
- Stephen M Cornish
- Faculty of Kinesiology and Recreation Management, University of Manitoba, 117 Frank Kennedy Centre, Winnipeg, Manitoba, R3T 2N2, Canada.
| | - Philip D Chilibeck
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Darren G Candow
- Faculty of Kinesiology and Health Studies, University of Regina, Regina, Saskatchewan, Canada
| |
Collapse
|
28
|
TLR7 Expression Is Associated with M2 Macrophage Subset in Calcific Aortic Valve Stenosis. Cells 2020; 9:cells9071710. [PMID: 32708790 PMCID: PMC7407122 DOI: 10.3390/cells9071710] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS) is a common age-related disease characterized by active calcification of the leaflets of the aortic valve. How innate immune cells are involved in disease pathogenesis is not clear. In this study we investigate the role of the pattern recognition receptor Toll-like receptor 7 (TLR7) in CAVS, especially in relation to macrophage subtype. Human aortic valves were used for mRNA expression analysis, immunofluorescence staining, or ex vivo tissue assays. Response to TLR7 agonist in primary macrophages and valvular interstitial cells (VICs) were investigated in vitro. In the aortic valve, TLR7 correlated with M2 macrophage markers on mRNA levels. Expression was higher in the calcified part compared with the intermediate and healthy parts. TLR7+ cells were co-stained with M2-type macrophage receptors CD163 and CD206. Ex vivo stimulation of valve tissue with the TLR7 ligand imiquimod significantly increased secretion of IL-10, TNF-α, and GM-CSF. Primary macrophages responded to imiquimod with increased secretion of IL-10 while isolated VICs did not respond. In summary, in human aortic valves TLR7 expression is associated with M2 macrophages markers. Ex vivo tissue challenge with TLR7 ligand led to secretion of immunomodulatory cytokine IL-10. These results connect TLR7 activation in CAVS to reduced inflammation and improved clearance.
Collapse
|
29
|
Maraux M, Gaillardet A, Gally A, Saas P, Cherrier T. Human primary neutrophil mRNA does not contaminate human resolving macrophage mRNA after efferocytosis. J Immunol Methods 2020; 483:112810. [PMID: 32592772 DOI: 10.1016/j.jim.2020.112810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 05/20/2020] [Accepted: 06/15/2020] [Indexed: 02/07/2023]
Abstract
The ingestion of apoptotic corpses by macrophages, a process called efferocytosis, is a crucial step in inflammation resolution, since it alters macrophage phenotype toward a pro-resolving profile to foil inflammation and to favor tissue repair. Up to now, the resolving macrophages remain poorly characterized, especially in humans. Global investigations, like RNA sequencing, would be very helpful to unravel some features of these elusive cells. Nonetheless, these inquiries may be challenging in a single-species model, since the fate of ingested mRNA remains unknown and may hinder any subsequent mRNA investigations in the phagocyte. A full human model consisting of primary human neutrophil and primary human monocyte-derived macrophage co-culture was set up several decades ago to mimic in vitro the efferocytosis process. However, to our knowledge, this model has not been characterized as a suitable model to perform global mRNA investigations. Indeed, the extent of ingested neutrophil mRNA contamination has not been assessed in resolving macrophages. This work answers to this crucial question. Indeed, based on the protocols presented in this article, we demonstrate that neutrophil mRNA is severely degraded and is not able to cross-contaminate resolving macrophage mRNA, contrary to apoptotic human peripheral blood derived mononuclear cell (PBMC) or apoptotic leukemic Jurkat cell mRNA. Moreover, this allogenic co-culture system does not favor neither neutrophil activation nor macrophage pro-inflammatory cytokine release. Collectively, we highlight that this model of primary human neutrophil and primary human monocyte-derived macrophage co-culture is the best model for mRNA investigations in human resolving macrophages to help improving our knowledge on these crucial cells.
Collapse
Affiliation(s)
- M Maraux
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, F-25000 Besançon, France
| | - A Gaillardet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, F-25000 Besançon, France
| | - A Gally
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, F-25000 Besançon, France
| | - P Saas
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, F-25000 Besançon, France
| | - T Cherrier
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, F-25000 Besançon, France.
| |
Collapse
|
30
|
Asadzadeh Z, Safarzadeh E, Safaei S, Baradaran A, Mohammadi A, Hajiasgharzadeh K, Derakhshani A, Argentiero A, Silvestris N, Baradaran B. Current Approaches for Combination Therapy of Cancer: The Role of Immunogenic Cell Death. Cancers (Basel) 2020; 12:E1047. [PMID: 32340275 PMCID: PMC7226590 DOI: 10.3390/cancers12041047] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/08/2020] [Accepted: 04/17/2020] [Indexed: 12/31/2022] Open
Abstract
Cell death resistance is a key feature of tumor cells. One of the main anticancer therapies is increasing the susceptibility of cells to death. Cancer cells have developed a capability of tumor immune escape. Hence, restoring the immunogenicity of cancer cells can be suggested as an effective approach against cancer. Accumulating evidence proposes that several anticancer agents provoke the release of danger-associated molecular patterns (DAMPs) that are determinants of immunogenicity and stimulate immunogenic cell death (ICD). It has been suggested that ICD inducers are two different types according to their various activities. Here, we review the well-characterized DAMPs and focus on the different types of ICD inducers and recent combination therapies that can augment the immunogenicity of cancer cells.
Collapse
Affiliation(s)
- Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (Z.A.); (S.S.); (K.H.); (A.D.)
| | - Elham Safarzadeh
- Department of Immunology and Microbiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil 5618985991, Iran;
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (Z.A.); (S.S.); (K.H.); (A.D.)
| | - Ali Baradaran
- Research & Development Lab, BSD Robotics, 4500 Brisbane, Australia;
| | - Ali Mohammadi
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark;
| | - Khalil Hajiasgharzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (Z.A.); (S.S.); (K.H.); (A.D.)
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (Z.A.); (S.S.); (K.H.); (A.D.)
| | | | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (Z.A.); (S.S.); (K.H.); (A.D.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran
| |
Collapse
|
31
|
Yamazaki M, Maruyama S, Abé T, Tsuneki M, Kato H, Izumi K, Tanuma JI, Cheng J, Saku T. Rac1-dependent phagocytosis of apoptotic cells by oral squamous cell carcinoma cells: A possible driving force for tumor progression. Exp Cell Res 2020; 392:112013. [PMID: 32320683 DOI: 10.1016/j.yexcr.2020.112013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 03/30/2020] [Accepted: 04/15/2020] [Indexed: 01/13/2023]
Abstract
Apoptotic cell death frequently occurs in human cancer tissues including oral squamous cell carcinoma (SCC), wherein apoptotic tumor cells are phagocytosed not only by macrophages but also by neighboring tumor cells. We previously reported that the engulfment of apoptotic SCC cells by neighboring SCC cells frequently occurs at the invading front. Therefore, we hypothesized that the phagocytosis of these apoptotic cells by tumor cells contributes to disease progression. Herein, using cultured oral SCC cells, we aimed to confirm whether tumor cells actually phagocytose apoptotic cells and to examine whether cellular activities are regulated by the phagocytosis of apoptotic cells. Co-culture experiments showed that living cells could ingest apoptotic cells into phagolysosomes. NSC23766, an inhibitor of Rac1, which is a key regulator of phagocytic cup formation in professional phagocytes, dramatically suppressed the phagocytosis of apoptotic cells by living cells. Additionally, cell migration and the secretion of DKK1, a tumor-promoting protein, were enhanced by co-culture with apoptotic cells, whereas NSC23766 inhibited these effects. These results show that tumor cells can actively phagocytose apoptotic neighbors in a Rac1-dependent manner and that such activity increases their migration. The regulation of apoptotic cell phagocytosis thus represents new directions for therapeutic intervention for oral cancer.
Collapse
Affiliation(s)
- Manabu Yamazaki
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Satoshi Maruyama
- Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, Niigata, Japan
| | - Tatsuya Abé
- Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Masayuki Tsuneki
- Oral Pathology Section, Department of Surgical Pathology, Niigata University Hospital, Niigata, Japan
| | - Hiroko Kato
- Division of Biomimetics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Kenji Izumi
- Division of Biomimetics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Jun-Ichi Tanuma
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Jun Cheng
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takashi Saku
- Division of Oral Pathology, Department of Tissue Regeneration and Reconstruction, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan; Fukuoka Dental College, Fukuoka, Japan
| |
Collapse
|
32
|
Sordi R, Bet ÂC, Della Justina AM, Ramos GC, Assreuy J. The apoptosis clearance signal phosphatidylserine inhibits leukocyte migration and promotes inflammation resolution in vivo. Eur J Pharmacol 2020; 877:173095. [PMID: 32246923 DOI: 10.1016/j.ejphar.2020.173095] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/18/2020] [Accepted: 03/31/2020] [Indexed: 12/19/2022]
Abstract
Previous studies have shown that phagocytosis of apoptotic cells can tune the macrophage phenotype and trigger the resolution of inflammation. This mechanism is largely dependent on the recognition of phosphatidylserine (PS) residues on the outer membrane of dying cells. Therefore, we sought to assess the effects of PS-containing liposomes (mimics of apoptotic cells) on the leukocyte migration profile during the inflammatory process in vivo. Inflammation was induced by carrageenan injection into air pouches created on the dorsal region of mice, as this model enables convenient access to the exudates for further investigation. Mice were treated with PBS, PS-containing or phosphatidylcholine (PC)-containing liposomes (10, 30 or 100 mg/kg intraperitoneally [i.p.]). Starting 8 h after carrageenan injection, the level of leukocyte infiltration was monitored over three days. The PS-containing, but not PC-containing, liposomes reduced the polymorphonuclear (PMN) and mononuclear (MN) leukocyte influx into the inflamed pouches in a dose-dependent fashion. Most notably, these effects could also be adoptively transferred; that is, they were also found in mice injected with a liposome-free peritoneal lavage obtained from the mice that had received the intraperitoneal PS-liposome treatment. The effect of treatment with the PS-induced soluble mediators (PS-ISMs) was found to be dependent on the presence of peritoneal macrophages and was susceptible to heat, trypsin degradation, and cycloheximide treatment. The PS-containing liposomes promoted the reduction of PMN leukocyte influx by triggering the release of anti-inflammatory autacoids with a proteinaceous nature that were produced de novo after PS exposure.
Collapse
Affiliation(s)
- Regina Sordi
- Department of Pharmacology, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Ângela Cristina Bet
- Department of Pharmacology, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Ana Maria Della Justina
- Department of Pharmacology, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Gustavo Campos Ramos
- Department of Pharmacology, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Jamil Assreuy
- Department of Pharmacology, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil.
| |
Collapse
|
33
|
Etienne J, Liu C, Skinner CM, Conboy MJ, Conboy IM. Skeletal muscle as an experimental model of choice to study tissue aging and rejuvenation. Skelet Muscle 2020; 10:4. [PMID: 32033591 PMCID: PMC7007696 DOI: 10.1186/s13395-020-0222-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/12/2020] [Indexed: 12/16/2022] Open
Abstract
Skeletal muscle is among the most age-sensitive tissues in mammal organisms. Significant changes in its resident stem cells (i.e., satellite cells, SCs), differentiated cells (i.e., myofibers), and extracellular matrix cause a decline in tissue homeostasis, function, and regenerative capacity. Based on the conservation of aging across tissues and taking advantage of the relatively well-characterization of the myofibers and associated SCs, skeletal muscle emerged as an experimental system to study the decline in function and maintenance of old tissues and to explore rejuvenation strategies. In this review, we summarize the approaches for understanding the aging process and for assaying the success of rejuvenation that use skeletal muscle as the experimental system of choice. We further discuss (and exemplify with studies of skeletal muscle) how conflicting results might be due to variations in the techniques of stem cell isolation, differences in the assays of functional rejuvenation, or deciding on the numbers of replicates and experimental cohorts.
Collapse
Affiliation(s)
- Jessy Etienne
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Chao Liu
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Colin M Skinner
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, Berkeley, CA, 94720-3220, USA.
| |
Collapse
|
34
|
Horst AK, Tiegs G, Diehl L. Contribution of Macrophage Efferocytosis to Liver Homeostasis and Disease. Front Immunol 2019; 10:2670. [PMID: 31798592 PMCID: PMC6868070 DOI: 10.3389/fimmu.2019.02670] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022] Open
Abstract
The clearance of apoptotic cells is pivotal for both maintaining tissue homeostasis and returning to homeostasis after tissue injury as part of the regenerative resolution response. The liver is known for its capacity to remove aged and damaged cells from the circulation and can serve as a graveyard for effector T cells. In particular Kupffer cells are active phagocytic cells, but during hepatic inflammatory responses incoming neutrophils and monocytes may contribute to pro-inflammatory damage. To stimulate resolution of such inflammation, myeloid cell function can change, via sensing of environmental changes in the inflammatory milieu. Also, the removal of apoptotic cells via efferocytosis and the signaling pathways that are activated in macrophages/phagocytes upon their engulfment of apoptotic cells are important for a return to tissue homeostasis. Here, we will discuss, how efferocytosis mechanisms in hepatic macrophages/phagocytes may regulate tissue homeostasis and be involved in tissue regeneration in liver disease.
Collapse
Affiliation(s)
- Andrea Kristina Horst
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gisa Tiegs
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Diehl
- Institute for Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
35
|
Han YK, Park GY, Bae MJ, Kim JS, Jo WS, Lee CG. Hypoxia induces immunogenic cell death of cancer cells by enhancing the exposure of cell surface calreticulin in an endoplasmic reticulum stress-dependent manner. Oncol Lett 2019; 18:6269-6274. [PMID: 31788104 DOI: 10.3892/ol.2019.10986] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 09/06/2019] [Indexed: 01/13/2023] Open
Abstract
Hypoxia is associated with resistance to anticancer therapies. Additionally, it is involved in the immune evasion of cancer cells by inducing an immunosuppressive microenvironment. However, the role of hypoxia in modulating the immunogenicity of cancer cells remains unknown. Hypoxia is known to induce endoplasmic reticulum (ER) stress, which serves a key role in inducing the cell surface exposure of calreticulin, a marker of immunogenic cell death. The present study investigated whether hypoxia influenced the immunogenicity of cancer cells using FACS, western blot analysis and syngenic mouse tumor model. The results revealed that hypoxia induced the cell surface exposure of calreticulin in human and mouse breast cancer cell lines depending on ER stress. Enhanced cell surface exposure of calreticulin induced by hypoxia resulted in an increase in anticancer immunity in a mouse model, which suggested that hypoxia induced immunogenic cell death. Notably, hypoxia did not significantly modulate the cell surface exposure of CD47, an antagonist of calreticulin function in cancer immunogenicity. These results suggest that hypoxia may enhance the immunogenicity of cancer cells themselves, in addition to its role in inducing an immunosuppressive cancer microenvironment.
Collapse
Affiliation(s)
- Yu Kyeong Han
- Department of Radiation Biology, Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea
| | - Ga-Young Park
- Department of Radiation Biology, Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea.,Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Min Ji Bae
- Department of Radiation Biology, Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea
| | - Joong Sun Kim
- K-Herbal Medicinal Research Center, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Wol Soon Jo
- Department of Radiation Biology, Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea
| | - Chang Geun Lee
- Department of Radiation Biology, Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan 46033, Republic of Korea
| |
Collapse
|
36
|
Desmonts de Lamache D, Moges R, Siddiq A, Allain T, Feener TD, Muench GP, McKenna N, Yates RM, Buret AG. Immuno-modulating properties of Tulathromycin in porcine monocyte-derived macrophages infected with porcine reproductive and respiratory syndrome virus. PLoS One 2019; 14:e0221560. [PMID: 31442273 PMCID: PMC6707645 DOI: 10.1371/journal.pone.0221560] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 08/09/2019] [Indexed: 01/04/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a positive-stranded RNA virus that grows in macrophages and causes acute pneumonia in pigs. PRRSV causes devastating losses to the porcine industry. However, due to its high antigenic variability and poorly understood immunopathogenesis, there is currently no effective vaccine or treatment to control PRRSV infection. The common occurrence of PRRSV infection with bacterial infections as well as its inflammatory-driven pathobiology raises the question of the value of antibiotics with immunomodulating properties for the treatment of the disease it causes. The macrolide antibiotic Tulathromycin (TUL) has been found to exhibit potent anti-inflammatory and immunomodulating properties in cattle and pigs. The aim of this study was to characterize the anti-viral and immunomodulating properties of TUL in PRRSV-infected porcine macrophages. Our findings indicate that blood monocyte-derived macrophages are readily infected by PRRSV and can be used as an effective cellular model to study PRRSV pathogenesis. TUL did not change intracellular or extracellular viral titers, not did it alter viral receptors (CD163 and CD169) expression on porcine macrophages. In contrast, TUL exhibited potent immunomodulating properties, which therefore occurred in the absence of any direct antiviral effects against PRRSV. TUL had an additive effect with PRRSV on the induction of macrophage apoptosis, and inhibited virus-induced necrosis. TUL significantly attenuated PRRSV-induced macrophage pro-inflammatory signaling (CXCL-8 and mitochondrial ROS production) and prevented PRRSV inhibition of non-opsonized and opsonized phagocytic function. Together, these data demonstrate that TUL inhibits PRRSV-induced inflammatory responses in porcine macrophages and protects against the phagocytic impairment caused by the virus. Research in live pigs is warranted to assess the potential clinical benefits of this antibiotic in the context of virally induced inflammation and tissue injury.
Collapse
Affiliation(s)
| | - R. Moges
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - A. Siddiq
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - T. Allain
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - T. D. Feener
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - G. P. Muench
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - N. McKenna
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
| | - R. M. Yates
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary AB, Canada
- Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, AB, Canada
| | - A. G. Buret
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- * E-mail:
| |
Collapse
|
37
|
Manoharan P, Song T, Radzyukevich TL, Sadayappan S, Lingrel JB, Heiny JA. KLF2 in Myeloid Lineage Cells Regulates the Innate Immune Response during Skeletal Muscle Injury and Regeneration. iScience 2019; 17:334-346. [PMID: 31326700 PMCID: PMC6652133 DOI: 10.1016/j.isci.2019.07.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/10/2019] [Accepted: 07/03/2019] [Indexed: 11/18/2022] Open
Abstract
Skeletal muscle repair and regeneration after injury requires coordinated interactions between the innate immune system and the injured muscle. Myeloid cells predominate in these interactions. This study examined the role of KLF2, a zinc-finger transcription factor that regulates immune cell activation, in specifying myeloid cell functions during muscle regeneration. Loss of KLF2 in myeloid lineage cells (myeKlf2-/- mice) dramatically enhanced the initial inflammatory response to acute muscle injury (cardiotoxin). Injured muscles showed dramatically elevated expression of inflammatory mediators and greater numbers of infiltrating, pro-inflammatory monocytes that matured earlier into activated macrophages. Notably, the inflammatory phase resolved earlier and regeneration progressed to myogenesis, marked by elevated expression of factors that promote the formation of new fibers from satellite cells. Regeneration was completed earlier, with phenotypically normal adult fibers integrated into the muscle syncytium. These findings identify myeloid KLF2 as a key regulator of myeloid cell functions in adult skeletal muscle regeneration.
Collapse
Affiliation(s)
- Palanikumar Manoharan
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA.
| | - Taejeong Song
- Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Tatiana L Radzyukevich
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Sakthivel Sadayappan
- Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Jerry B Lingrel
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA
| | - Judith A Heiny
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
38
|
Kim YB, Ahn YH, Jung JH, Lee YJ, Lee JH, Kang JL. Programming of macrophages by UV-irradiated apoptotic cancer cells inhibits cancer progression and lung metastasis. Cell Mol Immunol 2019; 16:851-867. [PMID: 30842627 PMCID: PMC6828747 DOI: 10.1038/s41423-019-0209-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 02/02/2019] [Indexed: 12/13/2022] Open
Abstract
Apoptotic cell clearance by phagocytes is essential in tissue homeostasis. We demonstrated that conditioned medium (CM) from macrophages exposed to apoptotic cancer cells inhibits the TGFβ1-induced epithelial–mesenchymal transition (EMT), migration, and invasion of cancer cells. Apoptotic 344SQ (ApoSQ) cell-induced PPARγ activity in macrophages increased the levels of PTEN, which was secreted in exosomes. Exosomal PTEN was taken up by recipient lung cancer cells. ApoSQ-exposed CM from PTEN knockdown cells failed to enhance PTEN in 344SQ cells, restore cellular polarity, or exert anti-EMT and anti-invasive effects. The CM that was deficient in PPARγ ligands, including 15-HETE, lipoxin A4, and 15d-PGJ2, could not reverse the suppression of PPARγ activity or the PTEN increase in 344SQ cells and consequently failed to prevent the EMT process. Moreover, a single injection of ApoSQ cells inhibited lung metastasis in syngeneic immunocompetent mice with enhanced PPARγ/PTEN signaling both in tumor-associated macrophages and in tumor cells. PPARγ antagonist GW9662 reversed the signaling by PPARγ/PTEN; the reduction in EMT-activating transcription factors, such as Snai1 and Zeb1; and the antimetastatic effect of the ApoSQ injection. Thus, the injection of apoptotic lung cancer cells may offer a new strategy for the prevention of lung metastasis.
Collapse
Affiliation(s)
- Yong-Bae Kim
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Young-Ho Ahn
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea.,Department of Molecular Medicine, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Ji-Hae Jung
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea.,Department of Physiology, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Ye-Ji Lee
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea.,Department of Physiology, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Jin-Hwa Lee
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea.,Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, 07804, Korea
| | - Jihee Lee Kang
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul, 07804, Korea. .,Department of Physiology, College of Medicine, Ewha Womans University, Seoul, 07804, Korea.
| |
Collapse
|
39
|
Szatmári-Tóth M, Ilmarinen T, Mikhailova A, Skottman H, Kauppinen A, Kaarniranta K, Kristóf E, Lytvynchuk L, Veréb Z, Fésüs L, Petrovski G. Human Embryonic Stem Cell-Derived Retinal Pigment Epithelium-Role in Dead Cell Clearance and Inflammation. Int J Mol Sci 2019; 20:ijms20040926. [PMID: 30791639 PMCID: PMC6412543 DOI: 10.3390/ijms20040926] [Citation(s) in RCA: 18] [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: 11/07/2018] [Revised: 12/19/2018] [Accepted: 02/13/2019] [Indexed: 12/19/2022] Open
Abstract
Inefficient removal of dying retinal pigment epithelial (RPE) cells by professional phagocytes can result in debris formation and development of age-related macular degeneration (AMD). Chronic oxidative stress and inflammation play an important role in AMD pathogenesis. Only a few well-established in vitro phagocytosis assay models exist. We propose human embryonic stem cell-derived-RPE cells as a new model for studying RPE cell removal by professional phagocytes. The characteristics of human embryonic stem cells-derived RPE (hESC-RPE) are similar to native RPEs based on their gene and protein expression profile, integrity, and barrier properties or regarding drug transport. However, no data exist about RPE death modalities and how efficiently dying hESC-RPEs are taken upby macrophages, and whether this process triggers an inflammatory responses. This study demonstrates hESC-RPEs can be induced to undergo anoikis or autophagy-associated cell death due to extracellular matrix detachment or serum deprivation and hydrogen-peroxide co-treatment, respectively, similar to primary human RPEs. Dying hESC-RPEs are efficiently engulfed by macrophages which results in high amounts of IL-6 and IL-8 cytokine release. These findings suggest that the clearance of anoikic and autophagy-associated dying hESC-RPEs can be used as a new model for investigating AMD pathogenesis or for testing the in vivo potential of these cells in stem cell therapy.
Collapse
Affiliation(s)
- Mária Szatmári-Tóth
- Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, 4032 Debrecen, Hungary.
| | - Tanja Ilmarinen
- Tampere University, Faculty of Medicine and Health Technology, 33014 Tampere, Finland.
| | - Alexandra Mikhailova
- Tampere University, Faculty of Medicine and Health Technology, 33014 Tampere, Finland.
| | - Heli Skottman
- Tampere University, Faculty of Medicine and Health Technology, 33014 Tampere, Finland.
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70211 Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70211 Kuopio, Finland.
- Department of Ophthalmology, Kuopio University Hospital, 70029 Kuopio, Finland.
| | - Endre Kristóf
- Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, 4032 Debrecen, Hungary.
| | - Lyubomyr Lytvynchuk
- Department of Ophthalmology, Justus-Liebig-University Giessen, Eye Clinic, University Hospital Giessen and Marburg GmbH, Campus Giessen, 35390 Giessen, Germany.
| | - Zoltán Veréb
- Department of Ophthalmology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
| | - László Fésüs
- Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, 4032 Debrecen, Hungary.
| | - Goran Petrovski
- Department of Biochemistry and Molecular Biology, University of Debrecen, Faculty of Medicine, 4032 Debrecen, Hungary.
- Department of Ophthalmology, Faculty of Medicine, University of Szeged, 6720 Szeged, Hungary.
- Center for Eye Research, Department of Ophthalmology, Oslo University Hospital and University of Oslo, Kirkeveien 166, 0450 Oslo, Norway.
| |
Collapse
|
40
|
Budai Z, Ujlaky-Nagy L, Kis GN, Antal M, Bankó C, Bacsó Z, Szondy Z, Sarang Z. Macrophages engulf apoptotic and primary necrotic thymocytes through similar phosphatidylserine-dependent mechanisms. FEBS Open Bio 2019; 9:446-456. [PMID: 30868053 PMCID: PMC6396166 DOI: 10.1002/2211-5463.12584] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/22/2018] [Accepted: 12/12/2018] [Indexed: 11/12/2022] Open
Abstract
One of the major roles of professional phagocytes is the removal of dead cells in the body. We know less about the clearance of necrotic cells than apoptotic cell phagocytosis, despite the fact that both types of dead cells need to be cleared together and necrotic cells appear often in pathological settings. In the present study, we examined phagocytosis of heat‐ or H2O2‐killed necrotic and apoptotic thymocytes by mouse bone marrow‐derived macrophages (BMDMs) in vitro and found that the two cell types are engulfed at equal efficiency and compete with each other when added together to BMDMs. Phagocytosis of both apoptotic and necrotic thymocytes was decreased by (a) blocking phosphatidylserine on the surface of dying cells; (b) inhibition of Mer tyrosine kinase, Tim‐4, integrin β3 receptor signaling, or Ras‐related C3 botulinum toxin substrate 1 activity; or (c) using BMDMs deficient for transglutaminase 2. Stimulation of liver X, retinoid X, retinoic acid or glucocorticoid nuclear receptors in BMDMs enhanced not only apoptotic, but also necrotic cell uptake. Electron microscopic analysis of the engulfment process revealed that the morphology of phagosomes and the phagocytic cup formed during the uptake of dying thymocytes is similar for apoptotic and necrotic cells. Our data indicate that apoptotic and necrotic cells are cleared via the same mechanisms, and removal of necrotic cells in vivo can be facilitated by molecules known to enhance the uptake of apoptotic cells.
Collapse
Affiliation(s)
- Zsófia Budai
- Department of Biochemistry and Molecular Biology Faculty of Medicine University of Debrecen Hungary
| | - László Ujlaky-Nagy
- Department of Biophysics and Cell Biology Faculty of Medicine University of Debrecen Hungary
| | - Gréta Nikoletta Kis
- Department of Anatomy, Histology and Embryology Faculty of Medicine University of Debrecen Hungary
| | - Miklós Antal
- Department of Anatomy, Histology and Embryology Faculty of Medicine University of Debrecen Hungary
| | - Csaba Bankó
- Department of Biophysics and Cell Biology Faculty of Medicine University of Debrecen Hungary
| | - Zsolt Bacsó
- Department of Biophysics and Cell Biology Faculty of Medicine and Faculty of Pharmacy University of Debrecen Hungary
| | - Zsuzsa Szondy
- Department of Biochemistry and Molecular Biology Faculty of Medicine University of Debrecen Hungary.,Department of Basic Medical Sciences Faculty of Dentistry University of Debrecen Hungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology Faculty of Medicine University of Debrecen Hungary
| |
Collapse
|
41
|
Miliani M, Nouar M, Paris O, Lefranc G, Mennechet F, Aribi M. Thymoquinone Potently Enhances the Activities of Classically Activated Macrophages Pulsed with Necrotic Jurkat Cell Lysates and the Production of Antitumor Th1-/M1-Related Cytokines. J Interferon Cytokine Res 2018; 38:539-551. [PMID: 30422744 DOI: 10.1089/jir.2018.0010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Antitumor activity of classically activated macrophage (Mϕ) may be impaired within the tumors, spleen, and bone marrow. Thus, it is possible to boost its antitumor activity after its pulsing with necrotic tumor cell lysates combined with an adjuvant. We set out to determine the potential adjuvant effects of thymoquinone (TQ; 2-isopropyl-5-methyl-1,4-benzoquinone, C10H12O2) on both functional activities of classically activated Mϕs, pulsed or not with necrotic Jurkat T cell line lysates (NecrJCL), and the balance of antitumor cytokines (ATCs) versus immunosuppressive cytokines (ISCs) during crosstalk with autologous human CD4+ T cells. We found that TQ treatment resulted in a significant upregulation of phagocytic activity, respiratory burst, the production of interleukin-2 (IL-2), IL-6, and IL-17 in NecrJCL-pulsed Mϕ co-culture system, and, conversely, in downregulation of the production of IL-6, IL-17, nitric oxide (NO), and arginase activity in nonpulsed TQ-treated Mϕs co-culture system. In addition, TQ has also shown low upregulation effect on the production of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and IL-1β, pathogen killing capacity and H2O2 in NecrJCL-pulsed Mϕs co-cultures. Moreover, TQ significantly downregulated arginase activity, and significantly upregulated inducible NO synthase (iNOS) activity-to-arginase activity ratio in NecrJCL-pulsed Mϕ co-cultures. Furthermore, TQ downregulated IL-10-to-IL-17 ratio and total cellular cholesterol content (ttcCHOL), but upregulated the ratios of IL-1β-to-IL-4, IL-1β-to-IL-10, IFN-γ-to-IL-4, IFN-γ-to-IL-10, TNF-α-to-IL-4, TNF-α-to-IL-10, and combined proinflammatory cytokines (PICs)-to-anti-inflammatory cytokines (AICs) in NecrJCL-pulsed Mϕs co-culture system, whereas significant differences were highlighted only for IL-10-to-IL-17, IFN-γ-to-IL-10, and PICs-to-AICs ratios. Our outcomes demonstrated that TQ can act as potent adjuvant for enhancing both the functional activities of NecrJCL-pulsed Mϕ and the production of ATCs during their interplay with CD4+ T cells.
Collapse
Affiliation(s)
- Maroua Miliani
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Mouna Nouar
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| | - Océane Paris
- Institut de Génétique Moléculaire de Montpellier (IGMM)-UMR5535, CNRS et Université de Montpellier, Montpellier, France
| | - Gérard Lefranc
- Institut de Génétique Humaine, UMR 9002 CNRS-Université de Montpellier, Montpellier, France
| | - Franck Mennechet
- Institut de Génétique Moléculaire de Montpellier (IGMM)-UMR5535, CNRS et Université de Montpellier, Montpellier, France
| | - Mourad Aribi
- Laboratory of Applied Molecular Biology and Immunology, W0414100, University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
42
|
Kumar N, Yadav N, Amarnath N, Sharma V, Shukla S, Srivastava A, Prasad P, Kumar A, Garg S, Singh S, Sehrawat S, Lochab B. Integrative natural medicine inspired graphene nanovehicle-benzoxazine derivatives as potent therapy for cancer. Mol Cell Biochem 2018; 454:123-138. [PMID: 30390174 DOI: 10.1007/s11010-018-3458-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 10/05/2018] [Indexed: 01/20/2023]
Abstract
Natural products from medicinal plants have always attracted a lot of attention due to their diverse and interesting therapeutic properties. We have employed the principles of green chemistry involving isomerization, coupling and condensation reaction to synthesize a class of compounds derived from eugenol, a naturally occurring bioactive phytophenol. The compounds were characterized structurally by 1H-, 13C-NMR, FT-IR spectroscopy and mass spectrometry analysis. The purity of compounds was detected by HPLC. The synthesized compounds exhibited anti-cancer activity. A 10-12-fold enhancement in efficiency of drug molecules (~ 1 µM) was observed when delivered with graphene oxide (GO) as a nanovehicle. Our data suggest cell death via apoptosis in a dose-dependent manner due to increase in calcium levels in specific cancer cell lines. Interestingly, the benzoxazine derivatives of eugenol with GO nanoparticle exhibited enhanced therapeutic potential in cancer cells. In addition to anti-cancer effect, we also observed significant role of these derivatives on parasite suggesting its multi-pharmacological capability.
Collapse
Affiliation(s)
- Naveen Kumar
- Brain Metastasis and NeuroVascular Disease Modelling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Nisha Yadav
- Materials Chemistry Lab, Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Nagarjuna Amarnath
- Materials Chemistry Lab, Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Vijeta Sharma
- Infectious Disease Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Swapnil Shukla
- Materials Chemistry Lab, Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Akriti Srivastava
- Infectious Disease Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Peeyush Prasad
- Brain Metastasis and NeuroVascular Disease Modelling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Anil Kumar
- James Graham Brown Cancer Centre, University of Louisville, 40202, Louisville, KY, USA
| | - Swati Garg
- Infectious Disease Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India
| | - Shailja Singh
- Infectious Disease Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India.
- Signaling Biology Lab, Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, India.
| | - Seema Sehrawat
- Brain Metastasis and NeuroVascular Disease Modelling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, NCR, India.
| | - Bimlesh Lochab
- Materials Chemistry Lab, Department of Chemistry, School of Natural Sciences, Shiv Nadar University, NCR, India.
| |
Collapse
|
43
|
Roy S, Bag AK, Dutta S, Polavaram NS, Islam R, Schellenburg S, Banwait J, Guda C, Ran S, Hollingsworth MA, Singh RK, Talmadge JE, Muders MH, Batra SK, Datta K. Macrophage-Derived Neuropilin-2 Exhibits Novel Tumor-Promoting Functions. Cancer Res 2018; 78:5600-5617. [PMID: 30111533 DOI: 10.1158/0008-5472.can-18-0562] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 06/06/2018] [Accepted: 08/07/2018] [Indexed: 12/14/2022]
Abstract
Tumor-associated macrophages (TAM) are causally associated with tumorigenesis as well as regulation of antitumor immune responses and have emerged as potential immunotherapeutic targets. Recent evidence suggests TAM phagocytose apoptotic tumor cells within the tumor microenvironment through efferocytosis in an immunologically silent manner, thus maintaining an immunosuppressed microenvironment. The signal transduction pathways coupling efferocytosis and immunosuppression are not well known. Neuropilin-2 (NRP2) is a member of the membrane-associated neuropilin family and has been reported in different immune cells but is poorly characterized. In this study, we show that NRP2 is expressed during macrophage differentiation, is induced by tumor cells, and regulates phagocytosis in macrophages. Furthermore, NRP2 in TAM promoted efferocytosis and facilitated tumor growth. Deletion of NRP2 from TAM impaired the clearance of apoptotic tumor cells and increased secondary necrosis within tumors. This resulted in a break in the immune tolerance and reinitiated antitumor immune responses, characterized by robust infiltration of CD8+ T and natural killer cells. This result suggests NRP2 may act as a molecular mediator that connects efferocytosis and immune suppression. Deletion of NRP2 in TAM downregulated several immunosuppressive and tumor-promoting genes and upregulated immunostimulatory genes in the myeloid compartment. Taken together, our study demonstrates that TAM-derived NRP2 plays a crucial role in tumor promotion through efferocytosis, opening the enticing option for the development of effective immunotherapy targeting TAM.Significance: Neuropilin-2 in macrophages promotes tumor growth by regulating efferocytosis of apoptotic tumor cells and orchestrating immune suppression.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/19/5600/F1.large.jpg Cancer Res; 78(19); 5600-17. ©2018 AACR.
Collapse
Affiliation(s)
- Sohini Roy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffet Cancer Center at University of Nebraska Medical Center, Omaha, Nebraska
| | - Arup K Bag
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffet Cancer Center at University of Nebraska Medical Center, Omaha, Nebraska
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffet Cancer Center at University of Nebraska Medical Center, Omaha, Nebraska
| | - Navatha Shree Polavaram
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffet Cancer Center at University of Nebraska Medical Center, Omaha, Nebraska
| | - Ridwan Islam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffet Cancer Center at University of Nebraska Medical Center, Omaha, Nebraska
| | - Samuel Schellenburg
- Institute of Pathology, University Hospital Carl Gustav Carus, University of Technology, Dresden, Germany
| | - Jasjit Banwait
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Chittibabu Guda
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sophia Ran
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois
| | - Michael A Hollingsworth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffet Cancer Center at University of Nebraska Medical Center, Omaha, Nebraska
- Department of Microbiology and Pathology, University of Nebraska Medical Center, Omaha, Nebraska
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska
| | - Rakesh K Singh
- Department of Microbiology and Pathology, University of Nebraska Medical Center, Omaha, Nebraska
| | - James E Talmadge
- Department of Microbiology and Pathology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michael H Muders
- Institute of Pathology, University Hospital Carl Gustav Carus, University of Technology, Dresden, Germany.
- Rudolf Becker Laboratory for Prostate Cancer Research, Center of Pathology, University of Bonn Medical Center, Bonn, Germany
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
- Fred and Pamela Buffet Cancer Center at University of Nebraska Medical Center, Omaha, Nebraska
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska.
- Fred and Pamela Buffet Cancer Center at University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
44
|
Sendler M, Weiss FU, Golchert J, Homuth G, van den Brandt C, Mahajan UM, Partecke LI, Döring P, Gukovsky I, Gukovskaya AS, Wagh PR, Lerch MM, Mayerle J. Cathepsin B-Mediated Activation of Trypsinogen in Endocytosing Macrophages Increases Severity of Pancreatitis in Mice. Gastroenterology 2018; 154:704-718.e10. [PMID: 29079517 PMCID: PMC6663074 DOI: 10.1053/j.gastro.2017.10.018] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 10/04/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Acute pancreatitis is characterized by premature intracellular activation of digestive proteases within pancreatic acini and a consecutive systemic inflammatory response. We investigated how these processes interact during severe pancreatitis in mice. METHODS Pancreatitis was induced in C57Bl/6 wild-type (control), cathepsin B (CTSB)-knockout, and cathepsin L-knockout mice by partial pancreatic duct ligation with supramaximal caerulein injection, or by repetitive supramaximal caerulein injections alone. Immune cells that infiltrated the pancreas were characterized by immunofluorescence detection of Ly6g, CD206, and CD68. Macrophages were isolated from bone marrow and incubated with bovine trypsinogen or isolated acinar cells; the macrophages were then transferred into pancreatitis control or cathepsin-knockout mice. Activities of proteases and nuclear factor (NF)-κB were determined using fluorogenic substrates and trypsin activity was blocked by nafamostat. Cytokine levels were measured using a cytometric bead array. We performed immunohistochemical analyses to detect trypsinogen, CD206, and CD68 in human chronic pancreatitis (n = 13) and acute necrotizing pancreatitis (n = 15) specimens. RESULTS Macrophages were the predominant immune cell population that migrated into the pancreas during induction of pancreatitis in control mice. CD68-positive macrophages were found to phagocytose acinar cell components, including zymogen-containing vesicles, in pancreata from mice with pancreatitis, as well as human necrotic pancreatic tissues. Trypsinogen became activated in macrophages cultured with purified trypsinogen or co-cultured with pancreatic acini and in pancreata of mice with pancreatitis; trypsinogen activation required macrophage endocytosis and expression and activity of CTSB, and was sensitive to pH. Activation of trypsinogen in macrophages resulted in translocation of NF-kB and production of inflammatory cytokines; mice without trypsinogen activation (CTSB-knockout mice) in macrophages developed less severe pancreatitis compared with control mice. Transfer of macrophage from control mice to CTSB-knockout mice increased the severity of pancreatitis. Inhibition of trypsin activity in macrophages prevented translocation of NF-κB and production of inflammatory cytokines. CONCLUSIONS Studying pancreatitis in mice, we found activation of digestive proteases to occur not only in acinar cells but also in macrophages that infiltrate pancreatic tissue. Activation of the proteases in macrophage occurs during endocytosis of zymogen-containing vesicles, and depends on pH and CTSB. This process involves macrophage activation via NF-κB-translocation, and contributes to systemic inflammation and severity of pancreatitis.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Cathepsin B/deficiency
- Cathepsin B/genetics
- Cathepsin B/metabolism
- Cathepsin L/deficiency
- Cathepsin L/genetics
- Cells, Cultured
- Ceruletide
- Coculture Techniques
- Cytokines/metabolism
- Disease Models, Animal
- Endocytosis
- Enzyme Activation
- Genetic Predisposition to Disease
- Humans
- Hydrogen-Ion Concentration
- Inflammation Mediators/metabolism
- Macrophages/enzymology
- Macrophages/immunology
- Macrophages/pathology
- Macrophages/transplantation
- Mice, Inbred C57BL
- Mice, Knockout
- NF-kappa B/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/deficiency
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Necrosis
- Pancreas/enzymology
- Pancreas/immunology
- Pancreas/pathology
- Pancreatectomy
- Pancreatitis, Acute Necrotizing/chemically induced
- Pancreatitis, Acute Necrotizing/enzymology
- Pancreatitis, Acute Necrotizing/immunology
- Pancreatitis, Acute Necrotizing/pathology
- Phagocytosis
- Phenotype
- Severity of Illness Index
- Time Factors
- Trypsinogen/metabolism
Collapse
Affiliation(s)
- Matthias Sendler
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Frank-Ulrich Weiss
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Janine Golchert
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Georg Homuth
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | | | - Ujjwal M Mahajan
- Medizinische Klinik und Poliklinik II, Universitätsklinikum der Ludwig-Maximilians-Universität, Klinikum Grosshadern, Munich, Germany
| | - Lars-Ivo Partecke
- Department of Surgery, University Medicine Greifswald, Greifswald, Germany
| | - Paula Döring
- Institute of Pathology, University Medicine Greifswald, Greifswald, Germany
| | - Ilya Gukovsky
- VA Greater Los Angeles Healthcare System; David Geffen School of Medicine, University of California at Los Angeles, California
| | - Anna S Gukovskaya
- VA Greater Los Angeles Healthcare System; David Geffen School of Medicine, University of California at Los Angeles, California
| | - Preshit R Wagh
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Markus M Lerch
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany
| | - Julia Mayerle
- Department of Medicine A, University Medicine Greifswald, Greifswald, Germany; Medizinische Klinik und Poliklinik II, Universitätsklinikum der Ludwig-Maximilians-Universität, Klinikum Grosshadern, Munich, Germany.
| |
Collapse
|
45
|
ATG-dependent phagocytosis in dendritic cells drives myelin-specific CD4 + T cell pathogenicity during CNS inflammation. Proc Natl Acad Sci U S A 2017; 114:E11228-E11237. [PMID: 29233943 DOI: 10.1073/pnas.1713664114] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Although reactivation and accumulation of autoreactive CD4+ T cells within the CNS are considered to play a key role in the pathogenesis of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), the mechanisms of how these cells recognize their target organ and induce sustained inflammation are incompletely understood. Here, we report that mice with conditional deletion of the essential autophagy protein ATG5 in classical dendritic cells (DCs), which are present at low frequencies in the nondiseased CNS, are completely resistant to EAE development following adoptive transfer of myelin-specific T cells and show substantially reduced in situ CD4+ T cell accumulation during the effector phase of the disease. Endogenous myelin peptide presentation to CD4+ T cells following phagocytosis of injured, phosphatidylserine-exposing oligodendroglial cells is abrogated in the absence of ATG5. Pharmacological inhibition of ATG-dependent phagocytosis by the cardiac glycoside neriifolin, an inhibitor of the Na+, K+-ATPase, delays the onset and reduces the clinical severity of EAE induced by myelin-specific CD4+ T cells. These findings link phagocytosis of injured oligodendrocytes, a pathological hallmark of MS lesions and during EAE, with myelin antigen processing and T cell pathogenicity, and identify ATG-dependent phagocytosis in DCs as a key regulator in driving autoimmune CD4+ T cell-mediated CNS damage.
Collapse
|
46
|
Kim YB, Yoon YS, Choi YH, Park EM, Kang JL. Interaction of macrophages with apoptotic cells inhibits transdifferentiation and invasion of lung fibroblasts. Oncotarget 2017; 8:112297-112312. [PMID: 29348826 PMCID: PMC5762511 DOI: 10.18632/oncotarget.22737] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 11/15/2017] [Indexed: 01/25/2023] Open
Abstract
The invasion of activated fibroblasts is a key mechanism of tissue fibrosis pathology. The recognition and uptake of apoptotic cells can induce the anti-fibrogenic programming of macrophages. We demonstrate that after interacting with apoptotic cells, macrophages secrete bioactive molecules that antagonize TGF-β1-induced increases in myofibroblast (fibroproliferative) phenotypic markers and reduce the enhanced invasive capacity of TGF-β1- or EGF-treated mouse lung fibroblasts (MLg). Furthermore, numerous treatment strategies prevented the anti-fibrotic effects of conditioned media, including transfection of macrophages with COX-2 or RhoA siRNAs or treatment of MLg cells with receptor antagonists for prostaglandin E2 (PGE2), PGD2, or hepatocyte growth factor (HGF). Additionally, administration of apoptotic cells in vivo inhibited the bleomycin-mediated invasive capacity of primary fibroblasts, as well as adhesion and extracellular matrix protein mRNA expression. These data suggest that the anti-fibrogenic programming of macrophages by apoptotic cells can be used as a novel tool to control the progressive fibrotic reaction.
Collapse
Affiliation(s)
- Yong-Bae Kim
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul 07985, Korea
| | - Young-So Yoon
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul 07985, Korea.,Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07985, Korea
| | - Youn-Hee Choi
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul 07985, Korea.,Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07985, Korea
| | - Eun-Mi Park
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul 07985, Korea.,Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul 07985, Korea
| | - Jihee Lee Kang
- Tissue Injury Defense Research Center, College of Medicine, Ewha Womans University, Seoul 07985, Korea.,Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07985, Korea
| |
Collapse
|
47
|
More Bayona JA, Karuppannan AK, Barreda DR. Contribution of leukocytes to the induction and resolution of the acute inflammatory response in chickens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 74:167-177. [PMID: 28456536 DOI: 10.1016/j.dci.2017.04.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 04/14/2017] [Accepted: 04/20/2017] [Indexed: 06/07/2023]
Abstract
A successful immune response against invading pathogens relies on the efficient activation of host defense mechanisms and a timely return to immune homeostasis. Despite their importance, these mechanisms remain ill-defined in most animal groups. This study focuses on the acute inflammatory response of chickens, important both as an avian model with a unique position in evolution as well as an increasingly notable target of infectious zoonotic diseases. We took advantage of an in vivo self-resolving intra-abdominal challenge model to provide an integrative view of leukocyte responses during the induction and resolution phases of acute inflammation. Our results showed rapid leukocyte infiltration into the abdominal cavity post zymosan challenge (significant increase as early as 4 h), which was dominated by heterophils. Peak leukocyte infiltration and ROS production reached maximum levels at 12 h post challenge, which was significantly earlier than comparative studies in teleost fish and mice. Both heterophils and monocyte/macrophages contributed to ROS production. Local leukocyte infiltration was preceded by an increase in peripheral leukocytes and a drop in the number of bone marrow leukocytes. The proportion of apoptotic leukocytes increased following peak of acute inflammation, rising to significant levels within the abdominal cavity by 48 h, consistent with other indicators for the resolution of inflammation. Importantly, comparison of chicken phagocytic responses with those previously shown in agnathan, teleost and murine models suggested a progressive evolutionary shift towards an increased sensitivity to pro-inflammatory pathogen-derived particles and decreased sensitivity towards homeostatic stimuli. Thus, while significant conservation can be noted across the immune systems of endotherms, this study highlights additional unique features that govern the induction and resolution of acute inflammation in the avian system, which may be relevant to disease susceptibility and performance.
Collapse
Affiliation(s)
- Juan A More Bayona
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Anbu Kumar Karuppannan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
| | - Daniel R Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta T6G 2P5, Canada; Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta T6G 2P5, Canada.
| |
Collapse
|
48
|
Abstract
The engulfment of apoptotic cells by phagocytes, a process referred to as efferocytosis, is essential for maintenance of normal tissue homeostasis and a prerequisite for the resolution of inflammation. Neutrophils are the predominant circulating white blood cell in humans, and contain an arsenal of toxic substances that kill and degrade microbes. Neutrophils are short-lived and spontaneously die by apoptosis. This review will highlight how the engulfment of apoptotic neutrophils by human phagocytes occurs, how heterogeneity of phagocyte populations influences efferocytosis signaling, and downstream consequences of efferocytosis. The efferocytosis of apoptotic neutrophils by macrophages promotes anti-inflammatory signaling, prevents neutrophil lysis, and dampens immune responses. Given the immunomodulatory properties of efferocytosis, understanding pathways that regulate and enhance efferocytosis could be harnessed to combat infection and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Mallary C Greenlee-Wacker
- Inflammation Program, Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine University of Iowa, Veterans Administration Medical Center, Iowa City, IA, USA
| |
Collapse
|
49
|
Abstract
Cutaneous wound repair is a complex, dynamic process with the goal of rapidly sealing any breach in the skin's protective barrier. Myeloid cells compose a significant proportion of the inflammatory cells recruited to a wound site and play important roles in decontaminating the injured tissue of any invading microorganisms. Subsequently, myeloid cells are able to influence many aspects of the healing response, in part through their capacity to release a large array of signaling molecules that allow them to communicate with and regulate the behavior of other wound cells and in turn, be themselves exquisitely regulated by the wound microenvironment. Macrophages, for example, appear to play important, temporally changing roles in the initiation of scarring and subsequently in matrix remodeling to resolve fibrosis. In this way, myeloid cells seem to play both positive (e.g., pathogen killing and matrix remodeling) and negative (e.g., scarring) roles in wound repair. Further research is of course needed to elucidate the precise temporal and spatial myeloid cell phenotypes and behaviors and ultimately to design effective strategies to optimize the beneficial functions of these cells while minimizing their detrimental contributions to improve wound healing in the clinic.
Collapse
|
50
|
Juban G, Chazaud B. Metabolic regulation of macrophages during tissue repair: insights from skeletal muscle regeneration. FEBS Lett 2017; 591:3007-3021. [PMID: 28555751 DOI: 10.1002/1873-3468.12703] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/19/2022]
Abstract
Macrophages are highly versatile cells that are involved both in the mounting and the resolution of inflammatory responses. Besides their properties in innate immunity to fight against pathogens, macrophages are essential for tissue repair, during which they adopt sequential inflammatory status. While the acquisition of some canonical polarized inflammatory statuses in vitro (M1/M2) is beginning to be understood at the molecular level, the regulation of macrophage skewing in vivo has been less investigated. Immunometabolism, in particular, is an emerging field, and most of the studies so far have investigated the control of macrophage polarization using in vitro set-ups. In this context, skeletal muscle regeneration is an excellent paradigm to study tissue repair, since the sequential steps of inflammatory response and tissue repair are well characterized. In this Review, after introducing macrophage populations and functions during skeletal muscle regeneration, we present the current knowledge on the metabolic regulation of macrophage inflammatory status, with particular emphasis on the comparison between in vitro and in vivo models of macrophage activation. We also discuss the metabolic regulation of macrophages in vivo during skeletal muscle regeneration.
Collapse
Affiliation(s)
- Gaëtan Juban
- INSERM U1217, CNRS 5310, Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Université de Lyon, Villeurbanne, France
| | - Bénédicte Chazaud
- INSERM U1217, CNRS 5310, Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Université de Lyon, Villeurbanne, France
| |
Collapse
|