451
|
Welz B, Bikker R, Hoffmeister L, Diekmann M, Christmann M, Brand K, Huber R. Activation of GSK3 Prevents Termination of TNF-Induced Signaling. J Inflamm Res 2021; 14:1717-1730. [PMID: 33986607 PMCID: PMC8111165 DOI: 10.2147/jir.s300806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/10/2021] [Indexed: 12/14/2022] Open
Abstract
Background Termination of TNF-induced signaling plays a key role in the resolution of inflammation with dysregulations leading to severe pathophysiological conditions (sepsis, chronic inflammatory disease, cancer). Since a recent phospho-proteome analysis in human monocytes suggested GSK3 as a relevant kinase during signal termination, we aimed at further elucidating its role in this context. Materials and Methods For the analyses, THP-1 monocytic cells and primary human monocytes were used. Staurosporine (Stauro) was applied to activate GSK3 by inhibiting kinases that mediate inhibitory GSK3α/β-Ser21/9 phosphorylation (eg, PKC). For GSK3 inhibition, Kenpaulone (Ken) was used. GSK3- and PKC-siRNAs were applied for knockdown experiments. Protein expression and phosphorylation were assessed by Western blot or ELISA and mRNA expression by qPCR. NF-κB activation was addressed using reporter gene assays. Results Constitutive GSK3β and PKCβ expression and GSK3α/β-Ser21/9 and PKCα/βII-Thr638/641 phosphorylation were not altered during TNF long-term incubation. Stauro-induced GSK3 activation (demonstrated by Bcl3 reduction) prevented termination of TNF-induced signaling as reflected by strongly elevated IL-8 expression (used as an indicator) following TNF long-term incubation. A similar increase was observed in TNF short-term-exposed cells, and this effect was inhibited by Ken. PKCα/β-knockdown modestly increased, whereas GSK3α/β-knockdown inhibited TNF-induced IL-8 expression. TNF-dependent activation of two NF-κB-dependent indicator plasmids was enhanced by Stauro, demonstrating transcriptional effects. A TNF-induced increase in p65-Ser536 phosphorylation was further enhanced by Stauro, whereas IκBα proteolysis and IKKα/β-Ser176/180 phosphorylation were not affected. Moreover, PKCβ-knockdown reduced levels of Bcl3. A20 and IκBα mRNA, both coding for signaling inhibitors, were dramatically less affected under our conditions when compared to IL-8, suggesting differential transcriptional effects. Conclusion Our results suggest that GSK3 activation is involved in preventing the termination of TNF-induced signaling. Our data demonstrate that activation of GSK3 – either pathophysiologically or pharmacologically induced – may destroy the finely balanced condition necessary for the termination of inflammation-associated signaling.
Collapse
Affiliation(s)
- Bastian Welz
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Rolf Bikker
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Leonie Hoffmeister
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Mareike Diekmann
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Martin Christmann
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - Korbinian Brand
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| | - René Huber
- Institute of Clinical Chemistry, Hannover Medical School, Hannover, 30625, Germany
| |
Collapse
|
452
|
Dai C, Me R, Gao N, Su G, Wu X, Yu FSX. Role of IL-36γ/IL-36R Signaling in Corneal Innate Defense Against Candida albicans Keratitis. Invest Ophthalmol Vis Sci 2021; 62:10. [PMID: 33970198 PMCID: PMC8114008 DOI: 10.1167/iovs.62.6.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Interleukin (IL)-36 cytokines have been shown to play either beneficial or detrimental roles in the infection of mucosal tissues in a pathogen-dependent manner, but their involvement in fungal keratitis remains elusive. We herein investigated their expression and function in mediating corneal innate immunity against Candida albicans infection. METHODS Gene expression in mouse corneas with or without C. albicans infection was determined by regular RT- and real-time (q)-PCR, Western blot analysis, ELISA or proteome profile assay. The severity of C. albicans keratitis was assessed using clinical scoring, bacterial counting, and myeloperoxidase (MPO) activity as an indicator of neutrophil infiltration. IL36R knockout mice and IL-33-specific siRNA were used to assess the involvement IL-33 signaling in C. albicans-infected corneas. B6 CD11c-DTR mice and clodronate liposomes were used to define the involvement of dendritic cells (DCs) and macrophages in IL-36R signaling and C. albicans keratitis, respectively. RESULTS IL-36γ were up-regulated in C57BL6 mouse corneas in response to C. albicans infection. IL-36 receptor-deficient mice display increased severity of keratitis, with a higher fungal load, MPO, and IL-1β levels, and lower soluble sIL-1Ra and calprotectin levels. Exogenous IL-36γ prevented fungal keratitis pathogenesis with lower fungal load and MPO activity, higher expression of sIL-1Ra and calprotectin, and lower expression of IL-1β, at mRNA or protein levels. Protein array analysis revealed that the expression of IL-33 and REG3G were related to IL-36/IL36R signaling, and siRNA downregulation of IL-33 increased the severity of C. albicans keratitis. Depletion of dendritic cells or macrophages resulted in severe C. albicans keratitis and yet exhibited minimal effects on exogenous IL-36γ-induced protection against C. albicans infection in B6 mouse corneas. CONCLUSIONS IL-36/IL36R signaling plays a protective role in fungal keratitis by promoting AMP expression and by suppressing fungal infection-induced expression of proinflammatory cytokines in a dendritic cell- and macrophage-independent manner.
Collapse
Affiliation(s)
- Chenyang Dai
- Departments of Ophthalmology and Anatomy and Cell Biology Wayne State University School of Medicine, Detroit, Michigan, United States
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Rao Me
- Departments of Ophthalmology and Anatomy and Cell Biology Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Nan Gao
- Departments of Ophthalmology and Anatomy and Cell Biology Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Guanyu Su
- Departments of Ophthalmology and Anatomy and Cell Biology Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Xinyi Wu
- Department of Ophthalmology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Fu-Shin X. Yu
- Departments of Ophthalmology and Anatomy and Cell Biology Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
453
|
Affiliation(s)
- Zachary W Wagoner
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA.,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA
| | - Weian Zhao
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA. .,Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, USA. .,Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA. .,Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA, USA. .,Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA. .,Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
454
|
Lee SA, Lee J, Kim K, Moon H, Min C, Moon B, Kim D, Yang S, Park H, Lee G, Park R, Park D. The Peroxisomal Localization of Hsd17b4 Is Regulated by Its Interaction with Phosphatidylserine. Mol Cells 2021; 44:214-222. [PMID: 33935042 PMCID: PMC8112170 DOI: 10.14348/molcells.2021.2217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/20/2021] [Accepted: 04/05/2021] [Indexed: 12/28/2022] Open
Abstract
Phosphatidylserine (PS), a negatively charged phospholipid exclusively located in the inner leaflet of the plasma membrane, is involved in various cellular processes such as blood coagulation, myoblast fusion, mammalian fertilization, and clearance of apoptotic cells. Proteins that specifically interact with PS must be identified to comprehensively understand the cellular processes involving PS. However, only a limited number of proteins are known to associate with PS. To identify PS-associating proteins, we performed a pulldown assay using streptavidin-coated magnetic beads on which biotin-linked PS was immobilized. Using this approach, we identified Hsd17b4, a peroxisomal protein, as a PS-associating protein. Hsd17b4 strongly associated with PS, but not with phosphatidylcholine or sphingomyelin, and the Scp-2-like domain of Hsd17b4 was responsible for this association. The association was disrupted by PS in liposomes, but not by free PS or the components of PS. In addition, translocation of PS to the outer leaflet of the plasma membrane enriched Hsd17b4 in peroxisomes. Collectively, this study suggests an unexpected role of PS as a regulator of the subcellular localization of Hsd17b4.
Collapse
Affiliation(s)
- Sang-Ah Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Juyeon Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Kwanhyeong Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Hyunji Moon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Chanhyuk Min
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Byeongjin Moon
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Deokhwan Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Susumin Yang
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Hyunjin Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Gwangrog Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, GIST, Gwangju 61005, Korea
| | - Daeho Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Center for Cell Mechanobiology, GIST, Gwangju 61005, Korea
| |
Collapse
|
455
|
Mike JK, Ferriero DM. Efferocytosis Mediated Modulation of Injury after Neonatal Brain Hypoxia-Ischemia. Cells 2021; 10:1025. [PMID: 33925299 PMCID: PMC8146813 DOI: 10.3390/cells10051025] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
Neonatal brain hypoxia-ischemia (HI) is a leading cause of morbidity and long-term disabilities in children. While we have made significant progress in describing HI mechanisms, the limited therapies currently offered for HI treatment in the clinical setting stress the importance of discovering new targetable pathways. Efferocytosis is an immunoregulatory and homeostatic process of clearance of apoptotic cells (AC) and cellular debris, best described in the brain during neurodevelopment. The therapeutic potential of stimulating defective efferocytosis has been recognized in neurodegenerative diseases. In this review, we will explore the involvement of efferocytosis after a stroke and HI as a promising target for new HI therapies.
Collapse
Affiliation(s)
- Jana Krystofova Mike
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Donna Marie Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA;
- Department of Neurology Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
456
|
Hughes LD, Wang Y, Meli AP, Rothlin CV, Ghosh S. Decoding Cell Death: From a Veritable Library of Babel to Vade Mecum? Annu Rev Immunol 2021; 39:791-817. [PMID: 33902311 DOI: 10.1146/annurev-immunol-102819-072601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Programmed cell death (PCD) is a requisite feature of development and homeostasis but can also be indicative of infections, injuries, and pathologies. In concordance with these heterogeneous contexts, an array of disparate effector responses occur downstream of cell death and its clearance-spanning tissue morphogenesis, homeostatic turnover, host defense, active dampening of inflammation, and tissue repair. This raises a fundamental question of how a single contextually appropriate response ensues after an event of PCD. To explore how complex inputs may together tailor the specificity of the resulting effector response, here we consider (a) the varying contexts during which different cell death modalities are observed, (b) the nature of the information that can be passed on by cell corpses, and (c) the ways by which efferocyte populations synthesize signals from dying cells with those from the surrounding microenvironment.
Collapse
Affiliation(s)
- Lindsey D Hughes
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; , , ,
| | - Yaqiu Wang
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; , , ,
| | - Alexandre P Meli
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; , , ,
| | - Carla V Rothlin
- Department of Immunobiology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; , , , .,Department of Pharmacology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA;
| | - Sourav Ghosh
- Department of Pharmacology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA; .,Department of Neurology, School of Medicine, Yale University, New Haven, Connecticut 06520, USA
| |
Collapse
|
457
|
Felix FB, Vago JP, Fernandes DDO, Martins DG, Moreira IZ, Gonçalves WA, Costa WC, Araújo JMD, Queiroz-Junior CM, Campolina-Silva GH, Soriani FM, Sousa LP, Grespan R, Teixeira MM, Pinho V. Biochanin A Regulates Key Steps of Inflammation Resolution in a Model of Antigen-Induced Arthritis via GPR30/PKA-Dependent Mechanism. Front Pharmacol 2021; 12:662308. [PMID: 33995086 PMCID: PMC8114065 DOI: 10.3389/fphar.2021.662308] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Biochanin A (BCA) is a natural organic compound of the class of phytochemicals known as flavonoids and isoflavone subclass predominantly found in red clover (Trifolium pratense). It has anti-inflammatory activity and some pro-resolving actions, such as neutrophil apoptosis. However, the effect of BCA in the resolution of inflammation is still poorly understood. In this study, we investigated the effects of BCA on the neutrophilic inflammatory response and its resolution in a model of antigen-induced arthritis. Male wild-type BALB/c mice were treated with BCA at the peak of the inflammatory process (12 h). BCA decreased the accumulation of migrated neutrophils, and this effect was associated with reduction of myeloperoxidase activity, IL-1β and CXCL1 levels, and the histological score in periarticular tissues. Joint dysfunction, as seen by mechanical hypernociception, was improved by treatment with BCA. The resolution interval (Ri) was also quantified, defining profiles of acute inflammatory parameters that include the amplitude and duration of the inflammatory response monitored by the neutrophil infiltration. BCA treatment shortened Ri from ∼23 h observed in vehicle-treated mice to ∼5.5 h, associated with an increase in apoptotic events and efferocytosis, both key steps for the resolution of inflammation. These effects of BCA were prevented by H89, an inhibitor of protein kinase A (PKA) and G15, a selective G protein–coupled receptor 30 (GPR30) antagonist. In line with the in vivo data, BCA also increased the efferocytic ability of murine bone marrow–derived macrophages. Collectively, these data indicate for the first time that BCA resolves neutrophilic inflammation acting in key steps of the resolution of inflammation, requiring activation of GPR30 and via stimulation of cAMP-dependent signaling.
Collapse
Affiliation(s)
- Franciel Batista Felix
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Priscila Vago
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Débora de Oliveira Fernandes
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Débora Gonzaga Martins
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabella Zaidan Moreira
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - William Antonio Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Walyson Coelho Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Celso Martins Queiroz-Junior
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | - Frederico Marianetti Soriani
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lirlândia Pires Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Renata Grespan
- Departamento de Fisiologia, Universidade Federal de Sergipe, São Cristovão, Brazil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
458
|
Yin C, Heit B. Cellular Responses to the Efferocytosis of Apoptotic Cells. Front Immunol 2021; 12:631714. [PMID: 33959122 PMCID: PMC8093429 DOI: 10.3389/fimmu.2021.631714] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/29/2021] [Indexed: 12/17/2022] Open
Abstract
The rapid and efficient phagocytic clearance of apoptotic cells, termed efferocytosis, is a critical mechanism in the maintenance of tissue homeostasis. Removal of apoptotic cells through efferocytosis prevents secondary necrosis and the resultant inflammation caused by the release of intracellular contents. The importance of efferocytosis in homeostasis is underscored by the large number of inflammatory and autoimmune disorders, including atherosclerosis and systemic lupus erythematosus, that are characterized by defective apoptotic cell clearance. Although mechanistically similar to the phagocytic clearance of pathogens, efferocytosis differs from phagocytosis in that it is immunologically silent and induces a tissue repair response. Efferocytes face unique challenges resulting from the internalization of apoptotic cells, including degradation of the apoptotic cell, dealing with the extra metabolic load imposed by the processing of apoptotic cell contents, and the coordination of an anti-inflammatory, pro-tissue repair response. This review will discuss recent advances in our understanding of the cellular response to apoptotic cell uptake, including trafficking of apoptotic cell cargo and antigen presentation, signaling and transcriptional events initiated by efferocytosis, the coordination of an anti-inflammatory response and tissue repair, unique cellular metabolic responses and the role of efferocytosis in host defense. A better understanding of how efferocytic cells respond to apoptotic cell uptake will be critical in unraveling the complex connections between apoptotic cell removal and inflammation resolution and maintenance of tissue homeostasis.
Collapse
Affiliation(s)
- Charles Yin
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Center for Human Immunology, Western University, London, ON, Canada
| | - Bryan Heit
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Center for Human Immunology, Western University, London, ON, Canada
- Robarts Research Institute, London, ON, Canada
| |
Collapse
|
459
|
Julian L, Naylor G, Wickman GR, Rath N, Castino G, Stevenson D, Bryson S, Munro J, McGarry L, Mullin M, Rice A, Del Río Hernández A, Olson MF. Defective apoptotic cell contractility provokes sterile inflammation, leading to liver damage and tumour suppression. eLife 2021; 10:e61983. [PMID: 33871359 PMCID: PMC8087448 DOI: 10.7554/elife.61983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 04/17/2021] [Indexed: 01/20/2023] Open
Abstract
Apoptosis is characterized by profound morphological changes, but their physiological purpose is unknown. To characterize the role of apoptotic cell contraction, ROCK1 was rendered caspase non-cleavable (ROCK1nc) by mutating aspartate 1113, which revealed that ROCK1 cleavage was necessary for forceful contraction and membrane blebbing. When homozygous ROCK1nc mice were treated with the liver-selective apoptotic stimulus of diethylnitrosamine, ROCK1nc mice had more profound liver damage with greater neutrophil infiltration than wild-type mice. Inhibition of the damage-associated molecular pattern protein HMGB1 or signalling by its cognate receptor TLR4 lowered neutrophil infiltration and reduced liver damage. ROCK1nc mice also developed fewer diethylnitrosamine-induced hepatocellular carcinoma (HCC) tumours, while HMGB1 inhibition increased HCC tumour numbers. Thus, ROCK1 activation and consequent cell contraction are required to limit sterile inflammation and damage amplification following tissue-scale cell death. Additionally, these findings reveal a previously unappreciated role for acute sterile inflammation as an efficient tumour-suppressive mechanism.
Collapse
Affiliation(s)
- Linda Julian
- Cancer Research United Kingdom Beatson Institute, Garscube EstateGlasgowUnited Kingdom
- Institute of Cancer Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Gregory Naylor
- Cancer Research United Kingdom Beatson Institute, Garscube EstateGlasgowUnited Kingdom
- Institute of Cancer Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Grant R Wickman
- Cancer Research United Kingdom Beatson Institute, Garscube EstateGlasgowUnited Kingdom
- Institute of Cancer Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Nicola Rath
- Cancer Research United Kingdom Beatson Institute, Garscube EstateGlasgowUnited Kingdom
| | - Giovanni Castino
- Department of Chemistry and Biology, Ryerson UniversityTorontoCanada
| | - David Stevenson
- Cancer Research United Kingdom Beatson Institute, Garscube EstateGlasgowUnited Kingdom
| | - Sheila Bryson
- Cancer Research United Kingdom Beatson Institute, Garscube EstateGlasgowUnited Kingdom
| | - June Munro
- Cancer Research United Kingdom Beatson Institute, Garscube EstateGlasgowUnited Kingdom
| | - Lynn McGarry
- Cancer Research United Kingdom Beatson Institute, Garscube EstateGlasgowUnited Kingdom
| | - Margaret Mullin
- Electron Microscopy Facility, School of Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Alistair Rice
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College LondonLondonUnited Kingdom
| | - Armandodel Del Río Hernández
- Cellular and Molecular Biomechanics Laboratory, Department of Bioengineering, Imperial College LondonLondonUnited Kingdom
| | - Michael F Olson
- Department of Chemistry and Biology, Ryerson UniversityTorontoCanada
| |
Collapse
|
460
|
I'm Infected, Eat Me! Innate Immunity Mediated by Live, Infected Cells Signaling To Be Phagocytosed. Infect Immun 2021; 89:IAI.00476-20. [PMID: 33558325 DOI: 10.1128/iai.00476-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Innate immunity against pathogens is known to be mediated by barriers to pathogen invasion, activation of complement, recruitment of immune cells, immune cell phagocytosis of pathogens, death of infected cells, and activation of the adaptive immunity via antigen presentation. Here, we propose and review evidence for a novel mode of innate immunity whereby live, infected host cells induce phagocytes to phagocytose the infected cell, thereby potentially reducing infection. We discuss evidence that host cells, infected by virus, bacteria, or other intracellular pathogens (i) release nucleotides and chemokines as find-me signals, (ii) expose on their surface phosphatidylserine and calreticulin as eat-me signals, (iii) release and bind opsonins to induce phagocytosis, and (iv) downregulate don't-eat-me signals CD47, major histocompatibility complex class I (MHC1), and sialic acid. As long as the pathogens of the host cell are destroyed within the phagocyte, then infection can be curtailed; if antigens from the pathogens are cross-presented by the phagocyte, then an adaptive response would also be induced. Phagocytosis of live infected cells may thereby mediate innate immunity.
Collapse
|
461
|
Shankman LS, Fleury ST, Evans WB, Penberthy KK, Arandjelovic S, Blumberg RS, Agaisse H, Ravichandran KS. Efferocytosis by Paneth cells within the intestine. Curr Biol 2021; 31:2469-2476.e5. [PMID: 33852873 DOI: 10.1016/j.cub.2021.03.055] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 01/12/2021] [Accepted: 03/16/2021] [Indexed: 12/15/2022]
Abstract
Apoptotic cells are quickly and efficiently engulfed and removed via the process of efferocytosis by either professional phagocytes, such as macrophages, or non-professional phagocytes, including epithelial cells.1,2 In addition to debris removal, a key benefit of efferocytosis is that phagocytes engulfing apoptotic cells release anti-inflammatory mediators3,4 that help reduce local tissue inflammation;5 conversely, accumulation of uncleared apoptotic cells predisposes to a pro-inflammatory tissue milieu.6-8 Due to their high proliferative capacity, intestinal epithelial cells (iECs) are sensitive to inflammation, irradiation, and chemotherapy-induced DNA damage, leading to apoptosis. Mechanisms of iEC death in the context of irradiation has been studied,9,10 but phagocytosis of dying iECs is poorly understood. Here, we identify an unexpected efferocytic role for Paneth cells, which reside in intestinal crypts and are linked to innate immunity and maintenance of the stem cell niche in the crypt.11,12 Through a series of studies spanning in vitro efferocytosis, ex vivo intestinal organoids ("enteroids"), and in vivo Cre-mediated deletion of Paneth cells, we show that Paneth cells mediate apoptotic cell uptake of dying neighbors. The relevance of Paneth-cell-mediated efferocytosis was revealed ex vivo and in mice after low-dose cesium-137 (137Cs) irradiation, mimicking radiation therapies given to cancer patients often causing significant apoptosis of iECs. These data advance a new concept that Paneth cells can act as phagocytes and identify another way in which Paneth cells contribute to the overall health of the intestine. These observations also have implications for individuals undergoing chemotherapy or chronic inflammatory bowel disease.
Collapse
Affiliation(s)
- Laura S Shankman
- Center for Cell Clearance, University of Virginia, Jeanette Lancaster Way, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology, and Cancer Biology, Jefferson Park Avenue, University of Virginia, Charlottesville, VA 22908, USA
| | - Samantha T Fleury
- Center for Cell Clearance, University of Virginia, Jeanette Lancaster Way, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology, and Cancer Biology, Jefferson Park Avenue, University of Virginia, Charlottesville, VA 22908, USA
| | - W Britt Evans
- Center for Cell Clearance, University of Virginia, Jeanette Lancaster Way, Charlottesville, VA 22908, USA
| | - Kristen K Penberthy
- Center for Cell Clearance, University of Virginia, Jeanette Lancaster Way, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology, and Cancer Biology, Jefferson Park Avenue, University of Virginia, Charlottesville, VA 22908, USA
| | - Sanja Arandjelovic
- Center for Cell Clearance, University of Virginia, Jeanette Lancaster Way, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology, and Cancer Biology, Jefferson Park Avenue, University of Virginia, Charlottesville, VA 22908, USA
| | - Richard S Blumberg
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Francis Street, Boston, MA 02115, USA
| | - Hervé Agaisse
- Department of Microbiology, Immunology, and Cancer Biology, Jefferson Park Avenue, University of Virginia, Charlottesville, VA 22908, USA
| | - Kodi S Ravichandran
- Center for Cell Clearance, University of Virginia, Jeanette Lancaster Way, Charlottesville, VA 22908, USA; Department of Microbiology, Immunology, and Cancer Biology, Jefferson Park Avenue, University of Virginia, Charlottesville, VA 22908, USA; VIB-UGent Center for Inflammation Research, Technologiepark 71, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium.
| |
Collapse
|
462
|
Williamson B, Pilla Reddy V. Blood retinal barrier and ocular pharmacokinetics: Considerations for the development of oncology drugs. Biopharm Drug Dispos 2021; 42:128-136. [PMID: 33759216 DOI: 10.1002/bdd.2276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/09/2021] [Accepted: 03/14/2021] [Indexed: 12/12/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are an example of targeted drug therapy to treat cancer while minimizing damage to healthy tissue. In contrast to traditional oncology drugs, the toxicity profile of targeted therapies is less well understood and can include severe ocular adverse events, which are among the most common toxicity reported by these therapeutics. Inhibition of Mer receptor tyrosine kinase (MERTK) promotes innate tumor immunity by decreasing M2-macrophage polarization and efferocytosis. This mechanism offers the opportunity for targeted immunotherapy to treat cancer; however, the ocular expression of MERTK increases the difficulty for developing a targeted drug due to toxicity concerns. In this article we review the pharmacokinetic (PK) parameters and in vitro absorption, distribution, metabolism, and excretion (ADME) assays available to evaluate ocular disposition and assess the relationship between clinical PK and reported ocular events for TKIs to allow backtranslation to preclinical models. Understanding the ocular disposition in the context of PK and safety remains an evolving area and is likely to be a key aspect of developing safe and efficacious oncology drugs, devoid of ocular toxicity.
Collapse
Affiliation(s)
- Beth Williamson
- Drug Metabolism and Pharmacokinetics, Early Oncology, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Venkatesh Pilla Reddy
- Modelling and Simulation, Early Oncology, Oncology R&D, AstraZeneca, Cambridge, UK.,Clinical Pharmacology and Quantitative Pharmacology, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
463
|
Abstract
A fundamental concept in immunology is that the innate immune system initiates or instructs downstream adaptive immune responses. Inflammasomes are central players in innate immunity to pathogens, but how inflammasomes shape adaptive immunity is complex and relatively poorly understood. Here we highlight recent work on the interplay between inflammasomes and adaptive immunity. We address how inflammasome-dependent release of cytokines and antigen activates, shapes or even inhibits adaptive immune responses. We consider how distinct tissue or cellular contexts may alter the effects of inflammasome activation on adaptive immunity and how this contributes to beneficial or detrimental outcomes in infectious diseases, cancer and autoimmunity. We aspire to provide a framework for thinking about inflammasomes and their connection to the adaptive immune response.
Collapse
|
464
|
Strzyz P. Epithelia arm up for dead-cell clearance. Nat Rev Mol Cell Biol 2021; 22:242-243. [PMID: 33594279 DOI: 10.1038/s41580-021-00351-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
465
|
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are clinical syndromes that cause significant mortality in clinical settings and morbidity among survivors accompanied by huge healthcare costs. Lung-resident cell dysfunction/death and neutrophil alveolitis accompanied by proteinous edema are the main pathological features of ALI/ARDS. While understanding of the mechanisms underlying ALI/ARDS pathogenesis is progressing and potential treatments such as statin therapy, nutritional strategies, and mesenchymal cell therapy are emerging, poor clinical outcomes in ALI/ARDS patients persist. Thus, a better understanding of lung-resident cell death and neutrophil alveolitis and their mitigation and clearance mechanisms may provide new therapeutic strategies to accelerate lung repair and improve outcomes in critically ill patients. Macrophages are required for normal tissue development and homeostasis as well as regulating tissue injury and repair through modulation of inflammation and other cellular processes. While macrophages mediate various functions, here we review recent dead cell clearance (efferocytosis) mechanisms mediated by these immune cells for maintaining tissue homeostasis after infectious and non-infectious lung injury.
Collapse
Affiliation(s)
- Patrick M Noone
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| | - Sekhar P Reddy
- Department of Pediatrics, College of Medicine, University of Illinois at Chicago, IL 60612, USA
- Department of Pathology, College of Medicine, University of Illinois at Chicago, IL 60612, USA
| |
Collapse
|
466
|
Marques ARA, Ramos C, Machado-Oliveira G, Vieira OV. Lysosome (Dys)function in Atherosclerosis-A Big Weight on the Shoulders of a Small Organelle. Front Cell Dev Biol 2021; 9:658995. [PMID: 33855029 PMCID: PMC8039146 DOI: 10.3389/fcell.2021.658995] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/11/2021] [Indexed: 12/15/2022] Open
Abstract
Atherosclerosis is a progressive insidious chronic disease that underlies most of the cardiovascular pathologies, including myocardial infarction and ischemic stroke. The malfunctioning of the lysosomal compartment has a central role in the etiology and pathogenesis of atherosclerosis. Lysosomes are the degradative organelles of mammalian cells and process endogenous and exogenous substrates in a very efficient manner. Dysfunction of these organelles and consequent inefficient degradation of modified low-density lipoproteins (LDL) and apoptotic cells in atherosclerotic lesions have, therefore, numerous deleterious consequences for cellular homeostasis and disease progression. Lysosome dysfunction has been mostly studied in the context of the inherited lysosomal storage disorders (LSDs). However, over the last years it has become increasingly evident that the consequences of this phenomenon are more far-reaching, also influencing the progression of multiple acquired human pathologies, such as neurodegenerative diseases, cancer, and cardiovascular diseases (CVDs). During the formation of atherosclerotic plaques, the lysosomal compartment of the various cells constituting the arterial wall is under severe stress, due to the tremendous amounts of lipoproteins being processed by these cells. The uncontrolled uptake of modified lipoproteins by arterial phagocytic cells, namely macrophages and vascular smooth muscle cells (VSMCs), is the initial step that triggers the pathogenic cascade culminating in the formation of atheroma. These cells become pathogenic "foam cells," which are characterized by dysfunctional lipid-laden lysosomes. Here, we summarize the current knowledge regarding the origin and impact of the malfunctioning of the lysosomal compartment in plaque cells. We further analyze how the field of LSD research may contribute with some insights to the study of CVDs, particularly how therapeutic approaches that target the lysosomes in LSDs could be applied to hamper atherosclerosis progression and associated mortality.
Collapse
Affiliation(s)
- André R A Marques
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Cristiano Ramos
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Gisela Machado-Oliveira
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Otília V Vieira
- iNOVA4Health, Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade NOVA de Lisboa, Lisbon, Portugal
| |
Collapse
|
467
|
Chen X, Deng Z, Feng J, Chang Q, Lu F, Yuan Y. Necroptosis in Macrophage Foam Cells Promotes Fat Graft Fibrosis in Mice. Front Cell Dev Biol 2021; 9:651360. [PMID: 33842478 PMCID: PMC8027326 DOI: 10.3389/fcell.2021.651360] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/04/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Fibrosis is a major grafting-related complication that leads to fat tissue dysfunction. Macrophage-induced inflammation is related to the development of fat tissue fibrosis. Necroptosis is a recently discovered pathway of programmed cell necrosis that results in severe inflammation and subsequent tissue fibrosis. Thus, in this study, we investigated the role of macrophage necroptosis in fat graft fibrosis and the underlying mechanisms. Methods: Fibrosis and necroptosis were investigated in mouse fat tissue before and after grafting. An in vitro “crown-like” structure (CLS) cell culture model was developed by co-culturing RAW 264.7 macrophages with apoptotic adipocytes to reproduce in vivo CLS macrophage-adipocyte interactions. Lipid uptake and necroptosis in CLS macrophages were analyzed using Oil-Red-O staining, western blotting, and immunofluorescence. RAW264.7 macrophages were cultured alone or with apoptotic adipocytes and treated with a necroptosis inhibitor (Nec-1 or GSK872) to explore the paracrine effect of necroptotic CLS macrophages on collagen synthesis in fibroblasts in vitro. Mice were treated with Nec-1 to analyze the effect of blocking necroptosis on fat graft fibrosis. Results: Fibrosis was increased after grafting in fat grafts of mice. Macrophages clustered around apoptotic adipocytes or large oil droplets to form a typical CLS in fibrotic depots. This was accompanied by formation and necroptosis of macrophage foam cells (MFCs) in CLSs. RAW 264.7 macrophages co-cultured with apoptotic adipocytes induced CLS formation in vitro, and lipid accumulation in CLS macrophages resulted in the formation and necroptosis of MFCs. Necroptosis of MFCs altered the expression of collagen I and VI in fibroblasts via a paracrine mechanism involving inflammatory cytokines/chemokines, which was reversed by GSK872 or Nec-1 treatment. Furthermore, treatment with Nec-1 ameliorated fat graft fibrosis in mice. Conclusion: Apoptotic adipocytes induced necroptosis of MFCs, and necroptosis of these cells activated collagen synthesis in fibroblasts via a paracrine mechanism. Inhibition of necroptosis in macrophages is a potential approach to prevent fibrosis in fat grafts.
Collapse
Affiliation(s)
- Xihang Chen
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zilong Deng
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jingwei Feng
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yi Yuan
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
468
|
Kotlyarov S. Participation of ABCA1 Transporter in Pathogenesis of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2021; 22:3334. [PMID: 33805156 PMCID: PMC8037621 DOI: 10.3390/ijms22073334] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the important medical and social problem. According to modern concepts, COPD is a chronic inflammatory disease, macrophages play a key role in its pathogenesis. Macrophages are heterogeneous in their functions, which is largely determined by their immunometabolic profile, as well as the features of lipid homeostasis, in which the ATP binding cassette transporter A1 (ABCA1) plays an essential role. The objective of this work is the analysis of the ABCA1 protein participation and the function of reverse cholesterol transport in the pathogenesis of COPD. The expression of the ABCA1 gene in lung tissues takes the second place after the liver, which indicates the important role of the carrier in lung function. The participation of the transporter in the development of COPD consists in provision of lipid metabolism, regulation of inflammation, phagocytosis, and apoptosis. Violation of the processes in which ABCA1 is involved may be a part of the pathophysiological mechanisms, leading to the formation of a heterogeneous clinical course of the disease.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| |
Collapse
|
469
|
Abstract
Thromboinflammation involves complex interactions between actors of inflammation and immunity and components of the hemostatic system, which are elicited upon infection or tissue injury. In this context, the interplay between platelets and innate immune cells has been intensively investigated. The ATP-gated P2X1 ion channel, expressed on both platelets and neutrophils is of particular interest. On platelets, this ion channel contributes to platelet activation and thrombosis, especially under high shear stress conditions of small arteries, whereas on neutrophils, it is involved in chemotaxis and in mitigating the activation of circulating cells. In vitro studies indicate that it may also be implicated in platelet-dependent immune responses during bacterial infection. More recently, in a mouse model of intestinal epithelial barrier disruption causing systemic inflammation, it has been reported that neutrophil P2X1 ion channel could play a protective role against exaggerated inflammation-associated thrombosis. This review will focus on this unique role of the ATP-gated P2X1 ion channel in thromboinflammation, highlighting possible implications and pointing to the need for further investigation of the role of P2X1 ion channels in the interplay between platelets and neutrophils during thrombus formation under various sterile or infectious inflammatory settings and in distinct vascular beds.
Collapse
Affiliation(s)
- Cécile Oury
- GIGA Cardiovascular Sciences, Laboratory of Cardiology, University of Liège, Liège, Belgium
| | - Odile Wéra
- GIGA Cardiovascular Sciences, Laboratory of Cardiology, University of Liège, Liège, Belgium
| |
Collapse
|
470
|
Chen W, Li L, Wang J, Zhang R, Zhang T, Wu Y, Wang S, Xing D. The ABCA1-efferocytosis axis: A new strategy to protect against atherosclerosis. Clin Chim Acta 2021; 518:1-8. [PMID: 33741356 DOI: 10.1016/j.cca.2021.02.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022]
Abstract
Atherosclerosis, a disease process characterized by lipid accumulation and inflammation, is the main cause of coronary heart disease (CHD) and myocardial infarction (MI). Efferocytosis involves the clearance of apoptotic cells by phagocytes. Successful engulfment triggers the release of anti-inflammatory cytokines to suppress atherosclerosis. ABCA1 is a key mediator of cholesterol efflux to apoA-I for the generation of HDL-C in reverse cholesterol transport (RCT). Intriguingly, ABCA1 promotes not only cholesterol efflux but also efferocytosis. ABCA1 promotes efferocytosis by regulating the release of "find-me" ligands, including LPC, and the exposure, release, and expression of "eat-me" ligands, including PtdSer, ANXA1, ANXA5, MEGF10, and GULP1. ABCA1 has a pathway similar to TG2, which is an "eat-me" ligand. ABCA1 has the highest known homology to ABCA7, which controls efferocytosis as the engulfment and processing ligand. In addition, ABCA1 can form several regulatory feedback axes with ANXA1, MEGF10, GULP1, TNFα, and IL-6. Therefore, ABCA1 is the central factor that links cholesterol efflux and apoptotic cell clearance. Several drugs have been studied or approved for apoptotic cell clearance, such as CD47 antibody and PD1-/PD-L1 antibody. In this article, we review the role and mechanism of action of ABCA1 in efferocytosis and focus on new insights into the ABCA1-efferocytosis axis and its potential as a novel therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Lu Li
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Jie Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Renshuai Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Tingting Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Yudong Wu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China.
| | - Shuai Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Medical Imaging, Radiotherapy Department of Affiliated Hospital, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
471
|
Reprograming of Tumor-Associated Macrophages in Breast Tumor-Bearing Mice under Chemotherapy by Targeting Heme Oxygenase-1. Antioxidants (Basel) 2021; 10:antiox10030470. [PMID: 33809707 PMCID: PMC8002331 DOI: 10.3390/antiox10030470] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor-associated macrophages (TAMs) represent one of the most abundant components of the tumor microenvironment and play important roles in tumor development and progression. TAMs display plasticity and functional heterogeneity as reflected by distinct phenotypic subsets. TAMs with an M1 phenotype have proinflammatory and anti-tumoral properties whereas M2-like TAMs exert anti-inflammatory and pro-tumoral functions. Tumor cell debris generated during chemotherapy can stimulate primary tumor growth and recurrence. According to our previous study, phagocytic engulfment of breast tumor cell debris by TAMs attenuated chemotherapeutic efficacy through the upregulation of heme oxygenase-1 (HO-1). To verify the impact of HO-1 upregulation on the profile of macrophage polarization during cytotoxic therapy, we utilized a syngeneic murine breast cancer (4T1) model in which tumor bearing mice were treated with paclitaxel (PTX). PTX treatment markedly downregulated the surface expression of the M1 marker CD86 in infiltrated TAMs. Notably, there were significantly more cytotoxic CD8+ T cells in tumors of mice treated with PTX plus the HO-1 inhibitor, zinc protophorphyrin IX (ZnPP) than in mice treated with PTX alone. Interestingly, the tumor-inhibiting efficacy of PTX and ZnPP co-treatment was abrogated when macrophages were depleted by clodronate liposomes. Macrophage depletion also decreased the intratumoral CD8+ T cell population and downregulated the expression of Cxcl9 and Cxcl10. The expression of the M1 phenotype marker, CD86 was higher in mice injected with PTX plus ZnPP than that in mice treated with PTX alone. Conversely, the PTX-induced upregulation of the M2 marker gene, Il10 in CD11b+ myeloid cells from 4T1 tumor-bearing mice treated was dramatically reduced by the administration of the HO-1 inhibitor. Genetic ablation of HO-1 abolished the inhibitory effect of 4T1 tumor cell debris on expression of M1 marker genes, Tnf and Il12b, in LPS-stimulated BMDMs. HO-1-deficient BMDMs exposed to tumor cell debris also exhibited a diminished expression of the M2 macrophage marker, CD206. These findings, taken all together, provide strong evidence that HO-1 plays a pivotal role in the transition of tumor-inhibiting M1-like TAMs to tumor-promoting M2-like ones during chemotherapy.
Collapse
|
472
|
Cell-cell fusions and cell-in-cell phenomena in healthy cells and cancer: Lessons from protists and invertebrates. Semin Cancer Biol 2021; 81:96-105. [PMID: 33713795 DOI: 10.1016/j.semcancer.2021.03.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/28/2021] [Accepted: 03/04/2021] [Indexed: 02/08/2023]
Abstract
Herein we analyze two special routes of the multinucleated cells' formation - the fusion of mononuclear cells and the formation of cell-in-cell structures - in the healthy tissues and in tumorigenesis. There are many theories of tumorigenesis based on the phenomenon of emergence of the hybrid cancer cells. We consider the phenomena, which are rarely mentioned in those theories: namely, cellularization of syncytium or coenocytes, and the reversible or irreversible somatogamy. The latter includes the short-term and the long-term vegetative (somatic) cells' fusions in the life cycles of unicellular organisms. The somatogamy and multinuclearity have repeatedly and independently emerged in various groups of unicellular eukaryotes. These phenomena are among dominant survival and biodiversity sustaining strategies in protists and we admit that they can likely play an analogous role in cancer cells.
Collapse
|
473
|
Deretic V. Autophagy in inflammation, infection, and immunometabolism. Immunity 2021; 54:437-453. [PMID: 33691134 PMCID: PMC8026106 DOI: 10.1016/j.immuni.2021.01.018] [Citation(s) in RCA: 473] [Impact Index Per Article: 118.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/05/2020] [Accepted: 01/25/2021] [Indexed: 12/21/2022]
Abstract
Autophagy is a quality-control, metabolic, and innate immunity process. Normative autophagy affects many cell types, including hematopoietic as well as non-hematopoietic, and promotes health in model organisms and humans. When autophagy is perturbed, this has repercussions on diseases with inflammatory components, including infections, autoimmunity and cancer, metabolic disorders, neurodegeneration, and cardiovascular and liver diseases. As a cytoplasmic degradative pathway, autophagy protects from exogenous hazards, including infection, and from endogenous sources of inflammation, including molecular aggregates and damaged organelles. The focus of this review is on the role of autophagy in inflammation, including type I interferon responses and inflammasome outputs, from molecules to immune cells. A special emphasis is given to the intersections of autophagy with innate immunity, immunometabolism, and functions of organelles such as mitochondria and lysosomes that act as innate immunity and immunometabolic signaling platforms.
Collapse
Affiliation(s)
- Vojo Deretic
- Autophagy Inflammation and Metabolism (AIM) Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA; Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
474
|
Boutet MA, Courties G, Nerviani A, Le Goff B, Apparailly F, Pitzalis C, Blanchard F. Novel insights into macrophage diversity in rheumatoid arthritis synovium. Autoimmun Rev 2021; 20:102758. [PMID: 33476818 DOI: 10.1016/j.autrev.2021.102758] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease affecting joints and causing progressive damage and disability. Macrophages are of critical importance in the initiation and perpetuation of synovitis in RA, they can function as antigen presenting cells leading to T-cell dependent B-cell activation, assume a variety of inflammatory cell states with the production of destructive cytokines, but also contribute to tissue homeostasis/repair. The recent development of high-throughput technologies, including bulk and single cells RNA-sequencing, has broadened our understanding of synovial cell diversity, and opened novel perspectives to the discovery of new potential therapeutic targets in RA. In this review, we will focus on the relationship between the synovial macrophage infiltration and clinical disease severity and response to treatment. We will then provide a state-of-the-art picture of the biological roles of synovial macrophages and distinct macrophage subsets described in RA. Finally, we will review the effects of approved conventional and biologic drugs on the synovial macrophage component and highlight the therapeutic potential of future strategies to re-program macrophage phenotypes in RA.
Collapse
Affiliation(s)
- Marie-Astrid Boutet
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Gabriel Courties
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France.
| | - Alessandra Nerviani
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Benoit Le Goff
- INSERM UMR1238, Bone Sarcoma and Remodelling of Calcified Tissues, Nantes University, Nantes, France; Rheumatology Department, Nantes University Hospital, Nantes, France.
| | | | - Costantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Frédéric Blanchard
- INSERM UMR1238, Bone Sarcoma and Remodelling of Calcified Tissues, Nantes University, Nantes, France.
| |
Collapse
|
475
|
Aaes TL, Vandenabeele P. The intrinsic immunogenic properties of cancer cell lines, immunogenic cell death, and how these influence host antitumor immune responses. Cell Death Differ 2021; 28:843-860. [PMID: 33214663 PMCID: PMC7937679 DOI: 10.1038/s41418-020-00658-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/19/2020] [Accepted: 10/22/2020] [Indexed: 01/30/2023] Open
Abstract
Modern cancer therapies often involve the combination of tumor-directed cytotoxic strategies and generation of a host antitumor immune response. The latter is unleashed by immunotherapies that activate the immune system generating a more immunostimulatory tumor microenvironment and a stronger tumor antigen-specific immune response. Studying the interaction between antitumor cytotoxic therapies, dying cancer cells, and the innate and adaptive immune system requires appropriate experimental tumor models in mice. In this review, we discuss the immunostimulatory and immunosuppressive properties of cancer cell lines commonly used in immunogenic cell death (ICD) studies being apoptosis or necroptosis. We will especially focus on the antigenic component of immunogenicity. While in several cancer cell lines the epitopes of endogenously expressed tumor antigens are known, these intrinsic epitopes are rarely determined in experimental apoptotic or necroptotic ICD settings. Instead by far the most ICD research studies investigate the antigenic response against exogenously expressed model antigens such as ovalbumin or retroviral epitopes (e.g., AH1). In this review, we will argue that the immune response against endogenous tumor antigens and the immunopeptidome profile of cancer cell lines affect the eventual biological readouts in the typical prophylactic tumor vaccination type of experiments used in ICD research, and we will propose additional methods involving immunopeptidome profiling, major histocompatibility complex molecule expression, and identification of tumor-infiltrating immune cells to document intrinsic immunogenicity following different cell death modalities.
Collapse
Affiliation(s)
- Tania Løve Aaes
- grid.11486.3a0000000104788040Unit for Cell Clearance in Health and Disease, VIB Center for Inflammation Research, Ghent, Belgium ,grid.5342.00000 0001 2069 7798Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium ,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Peter Vandenabeele
- grid.5342.00000 0001 2069 7798Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium ,Cancer Research Institute Ghent (CRIG), Ghent, Belgium ,grid.11486.3a0000000104788040Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
476
|
Post SM, Andreeff M, DiNardo C, Khoury JD, Ruvolo PP. TAM kinases as regulators of cell death. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118992. [PMID: 33647320 DOI: 10.1016/j.bbamcr.2021.118992] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 01/16/2023]
Abstract
Receptor Tyrosine Kinases are critical regulators of signal transduction that support cell survival, proliferation, and differentiation. Dysregulation of normal Receptor Tyrosine Kinase function by mutation or other activity-altering event can be oncogenic or can impact the transformed malignant cell so it becomes particularly resistant to stress challenge, have increased proliferation, become evasive to immune surveillance, and may be more prone to metastasis of the tumor to other organ sites. The TAM family of Receptor Tyrosine Kinases (TYRO3, AXL, MERTK) is emerging as important components of malignant cell survival in many cancers. The TAM kinases are important regulators of cellular homeostasis and proper cell differentiation in normal cells as receptors for their ligands GAS6 and Protein S. They also are critical to immune and inflammatory processes. In malignant cells, the TAM kinases can act as ligand independent co-receptors to mutant Receptor Tyrosine Kinases and in some cases (e.g. FLT3-ITD mutant) are required for their function. They also have a role in immune checkpoint surveillance. At the time of this review, the Covid-19 pandemic poses a global threat to world health. TAM kinases play an important role in host response to many viruses and it is suggested the TAM kinases may be important in aspects of Covid-19 biology. This review will cover the TAM kinases and their role in these processes.
Collapse
Affiliation(s)
- Sean M Post
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America; Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Courtney DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Joseph D Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Peter P Ruvolo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America; Section of Molecular Hematology and Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America.
| |
Collapse
|
477
|
Green DR. Health and Fitness at the Single-Cell Level. Cancer Immunol Res 2021; 9:130-135. [PMID: 33536268 DOI: 10.1158/2326-6066.cir-20-0418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/14/2020] [Accepted: 11/18/2020] [Indexed: 11/16/2022]
Abstract
Genetically identical cells in a tissue can respond differently to perturbations in their environment or "stress." Such stresses can be physicochemical, mechanical, or infectious or may come from competition with other cells in the tissue. Here, I discuss how the varying responses to stress influence the decision of a cell to repair or die, and how one cell's response can have effects on surrounding cells. Such responses control the health and fitness of single cells and how they compete with other genetically identical cells.See related article on p. 129.
Collapse
Affiliation(s)
- Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
478
|
Rocchi C, Barazzuol L, Coppes RP. The evolving definition of salivary gland stem cells. NPJ Regen Med 2021; 6:4. [PMID: 33526786 PMCID: PMC7851389 DOI: 10.1038/s41536-020-00115-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 12/03/2020] [Indexed: 02/07/2023] Open
Abstract
Dysfunction of the salivary gland and irreversible hyposalivation are the main side effects of radiotherapy treatment for head and neck cancer leading to a drastic decrease of the quality of life of the patients. Approaches aimed at regenerating damaged salivary glands have been proposed as means to provide long-term restoration of tissue function in the affected patients. In studies to elucidate salivary gland regenerative mechanisms, more and more evidence suggests that salivary gland stem/progenitor cell behavior, like many other adult tissues, does not follow that of the hard-wired professional stem cells of the hematopoietic system. In this review, we provide evidence showing that several cell types within the salivary gland epithelium can serve as stem/progenitor-like cells. While these cell populations seem to function mostly as lineage-restricted progenitors during homeostasis, we indicate that upon damage specific plasticity mechanisms might be activated to take part in regeneration of the tissue. In light of these insights, we provide an overview of how recent developments in the adult stem cell research field are changing our thinking of the definition of salivary gland stem cells and their potential plasticity upon damage. These new perspectives may have important implications on the development of new therapeutic approaches to rescue radiation-induced hyposalivation.
Collapse
Affiliation(s)
- Cecilia Rocchi
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands. .,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands. .,Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, 5 Little France Drive, Edinburgh, EH16 4UU, UK.
| | - Lara Barazzuol
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| | - Rob P Coppes
- Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, University of Groningen, 9713 AV, Groningen, The Netherlands.,Department of Radiation Oncology, University Medical Center Groningen, University of Groningen, 9700 RB, Groningen, The Netherlands
| |
Collapse
|
479
|
Abstract
ABSTRACT As an integral component of cardiac tissue, macrophages are critical for cardiac development, adult heart homeostasis, as well as cardiac healing. One fundamental function of macrophages involves the clearance of dying cells or debris, a process termed efferocytosis. Current literature primarily pays attention to the impact of efferocytosis on apoptotic cells. However, emerging evidence suggests that necrotic cells and their released cellular debris can also be removed by cardiac macrophages through efferocytosis. Importantly, recent studies have demonstrated that macrophage efferocytosis plays an essential role in cardiac pathophysiology and repair. Therefore, understanding macrophage efferocytosis would provide valuable insights on cardiac health, and may offer new therapeutic strategies for the treatment of patients with heart failure. In this review, we first summarize the molecular signals that are associated with macrophage efferocytosis of apoptotic and necrotic cells, and then discuss how the linkage of efferocytosis to the resolution of inflammation affects cardiac function and recovery under normal and diseased conditions. Lastly, we highlight new discoveries related to the effects of macrophage efferocytosis on cardiac injury and repair.
Collapse
Affiliation(s)
- Li Yutian
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Li Qianqian
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Pharmaceutical Science, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, Ohio
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
480
|
Gastrin, via activation of PPARα, protects the kidney against hypertensive injury. Clin Sci (Lond) 2021; 135:409-427. [PMID: 33458737 DOI: 10.1042/cs20201340] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/05/2021] [Accepted: 01/15/2021] [Indexed: 12/16/2022]
Abstract
Hypertensive nephropathy (HN) is a common cause of end-stage renal disease with renal fibrosis; chronic kidney disease is associated with elevated serum gastrin. However, the relationship between gastrin and renal fibrosis in HN is still unknown. We, now, report that mice with angiotensin II (Ang II)-induced HN had increased renal cholecystokinin receptor B (CCKBR) expression. Knockout of CCKBR in mice aggravated, while long-term subcutaneous infusion of gastrin ameliorated the renal injury and interstitial fibrosis in HN and unilateral ureteral obstruction (UUO). The protective effects of gastrin on renal fibrosis can be independent of its regulation of blood pressure, because in UUO, gastrin decreased renal fibrosis without affecting blood pressure. Gastrin treatment decreased Ang II-induced renal tubule cell apoptosis, reversed Ang II-mediated inhibition of macrophage efferocytosis, and reduced renal inflammation. A screening of the regulatory factors of efferocytosis showed involvement of peroxisome proliferator-activated receptor α (PPAR-α). Knockdown of PPAR-α by shRNA blocked the anti-fibrotic effect of gastrin in vitro in mouse renal proximal tubule cells and macrophages. Immunofluorescence microscopy, Western blotting, luciferase reporter, and Cut&tag-qPCR analyses showed that CCKBR may be a transcription factor of PPAR-α, because gastrin treatment induced CCKBR translocation from cytosol to nucleus, binding to the PPAR-α promoter region, and increasing PPAR-α gene transcription. In conclusion, gastrin protects against HN by normalizing blood pressure, decreasing renal tubule cell apoptosis, and increasing macrophage efferocytosis. Gastrin-mediated CCKBR nuclear translocation may make it act as a transcription factor of PPAR-α, which is a novel signaling pathway. Gastrin may be a new potential drug for HN therapy.
Collapse
|
481
|
Koren E, Fuchs Y. Modes of Regulated Cell Death in Cancer. Cancer Discov 2021; 11:245-265. [PMID: 33462123 DOI: 10.1158/2159-8290.cd-20-0789] [Citation(s) in RCA: 234] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/15/2020] [Accepted: 10/29/2020] [Indexed: 11/16/2022]
Abstract
Cell suicide pathways, termed regulated cell death (RCD), play a critical role in organismal development, homeostasis, and pathogenesis. Here, we provide an overview of key RCD modalities, namely apoptosis, entosis, necroptosis, pyroptosis, and ferroptosis. We explore how various RCD modules serve as a defense mechanism against the emergence of cancer as well as the manner in which they can be exploited to drive oncogenesis. Furthermore, we outline current therapeutic agents that activate RCD and consider novel RCD-based strategies for tumor elimination. SIGNIFICANCE: A variety of antitumor therapeutics eliminate cancer cells by harnessing the devastating potential of cellular suicide pathways, emphasizing the critical importance of RCD in battling cancer. This review supplies a mechanistic perspective of distinct RCD modalities and explores the important role they play in tumorigenesis. We discuss how RCD modules serve as a double-edged sword as well as novel approaches aimed at selectively manipulating RCD for tumor eradication.
Collapse
Affiliation(s)
- Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel. Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel. Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
482
|
Rosenbaum SR, Wilski NA, Aplin AE. Fueling the Fire: Inflammatory Forms of Cell Death and Implications for Cancer Immunotherapy. Cancer Discov 2021; 11:266-281. [PMID: 33451983 DOI: 10.1158/2159-8290.cd-20-0805] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/04/2020] [Accepted: 10/01/2020] [Indexed: 11/16/2022]
Abstract
Unleashing the immune system with immune checkpoint inhibitors (ICI) has significantly improved overall survival for subsets of patients with stage III/IV cancer. However, many tumors are nonresponsive to ICIs, in part due to a lack of tumor-infiltrating lymphocytes (TIL). Converting these immune "cold" tumors to "hot" tumors that are thus more likely to respond to ICIs is a major obstacle for cancer treatment. Triggering inflammatory forms of cell death, such as necroptosis and pyroptosis, may alter the tumor immune microenvironment and the influx of TILs. We present an emerging view that promoting tumor-localized necroptosis and pyroptosis may ultimately enhance responses to ICI. SIGNIFICANCE: Many tumor types respond poorly to ICIs or respond but subsequently acquire resistance. Effective therapies for ICI-nonresponsive tumors are lacking and should be guided by evidence from preclinical studies. Promoting inflammatory cell death mechanisms within the tumor may alter the local immune microenvironment toward an ICI-responsive state.
Collapse
Affiliation(s)
- Sheera R Rosenbaum
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Nicole A Wilski
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. .,Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
483
|
Dinparastisaleh R, Mirsaeidi M. Antifibrotic and Anti-Inflammatory Actions of α-Melanocytic Hormone: New Roles for an Old Player. Pharmaceuticals (Basel) 2021; 14:ph14010045. [PMID: 33430064 PMCID: PMC7827684 DOI: 10.3390/ph14010045] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/16/2022] Open
Abstract
The melanocortin system encompasses melanocortin peptides, five receptors, and two endogenous antagonists. Besides pigmentary effects generated by α-Melanocytic Hormone (α-MSH), new physiologic roles in sexual activity, exocrine secretion, energy homeostasis, as well as immunomodulatory actions, exerted by melanocortins, have been described recently. Among the most common and burdensome consequences of chronic inflammation is the development of fibrosis. Depending on the regenerative capacity of the affected tissue and the quality of the inflammatory response, the outcome is not always perfect, with the development of some fibrosis. Despite the heterogeneous etiology and clinical presentations, fibrosis in many pathological states follows the same path of activation or migration of fibroblasts, and the differentiation of fibroblasts to myofibroblasts, which produce collagen and α-SMA in fibrosing tissue. The melanocortin agonists might have favorable effects on the trajectories leading from tissue injury to inflammation, from inflammation to fibrosis, and from fibrosis to organ dysfunction. In this review we briefly summarized the data on structure, receptor signaling, and anti-inflammatory and anti-fibrotic properties of α-MSH and proposed that α-MSH analogues might be promising future therapeutic candidates for inflammatory and fibrotic diseases, regarding their favorable safety profile.
Collapse
Affiliation(s)
- Roshan Dinparastisaleh
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, MD 21218, USA;
| | - Mehdi Mirsaeidi
- Division of Pulmonary and Critical Care, University of Miami, Miami, FL 33146, USA
- Correspondence: ; Tel.: +1-305-243-1377
| |
Collapse
|
484
|
The Seminiferous Epithelial Cycle of Spermatogenesis: Role of Non-receptor Tyrosine Kinases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1288:1-20. [PMID: 34453729 DOI: 10.1007/978-3-030-77779-1_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Non-receptor tyrosine kinases (NRTKs) are implicated in various biological processes including cell proliferation, differentiation, survival, and apoptosis, as well as cell adhesion and movement. NRTKs are expressed in all mammals and in different cell types, with extraordinarily high expression in the testis. Their association with the plasma membrane and dynamic subcellular localization are crucial parameters in their activation and function. Many NRTKs are found in endosomal protein trafficking pathways, which suggests a novel mechanism to regulate the timely junction restructuring in the mammalian testis to facilitate spermiation and germ cell transport across the seminiferous epithelium.
Collapse
|
485
|
Hertzog J, Rehwinkel J. Regulation and inhibition of the DNA sensor cGAS. EMBO Rep 2020; 21:e51345. [PMID: 33155371 PMCID: PMC7726805 DOI: 10.15252/embr.202051345] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/01/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022] Open
Abstract
Cell-autonomous sensing of nucleic acids is essential for host defence against invading pathogens by inducing antiviral and inflammatory cytokines. cGAS has emerged in recent years as a non-redundant DNA sensor important for detection of many viruses and bacteria. Upon binding to DNA, cGAS synthesises the cyclic dinucleotide 2'3'-cGAMP that binds to the adaptor protein STING and thereby triggers IRF3- and NFκB-dependent transcription. In addition to infection, the pathophysiology of an ever-increasing number of sterile inflammatory conditions in humans involves the recognition of DNA through cGAS. Consequently, the cGAS/STING signalling axis has emerged as an attractive target for pharmacological modulation. However, the development of cGAS and STING inhibitors has just begun and a need for specific and effective compounds persists. In this review, we focus on cGAS and explore how its activation by immunostimulatory DNA is regulated by cellular mechanisms, viral immune modulators and small molecules. We further use our knowledge of cGAS modulation by cells and viruses to conceptualise potential new ways of pharmacological cGAS targeting.
Collapse
Affiliation(s)
- Jonny Hertzog
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular MedicineRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Jan Rehwinkel
- MRC Human Immunology UnitMRC Weatherall Institute of Molecular MedicineRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
486
|
Kalinski AL, Yoon C, Huffman LD, Duncker PC, Kohen R, Passino R, Hafner H, Johnson C, Kawaguchi R, Carbajal KS, Jara JS, Hollis E, Geschwind DH, Segal BM, Giger RJ. Analysis of the immune response to sciatic nerve injury identifies efferocytosis as a key mechanism of nerve debridement. eLife 2020; 9:60223. [PMID: 33263277 PMCID: PMC7735761 DOI: 10.7554/elife.60223] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 12/01/2020] [Indexed: 12/12/2022] Open
Abstract
Sciatic nerve crush injury triggers sterile inflammation within the distal nerve and axotomized dorsal root ganglia (DRGs). Granulocytes and pro-inflammatory Ly6Chigh monocytes infiltrate the nerve first and rapidly give way to Ly6Cnegative inflammation-resolving macrophages. In axotomized DRGs, few hematogenous leukocytes are detected and resident macrophages acquire a ramified morphology. Single-cell RNA-sequencing of injured sciatic nerve identifies five macrophage subpopulations, repair Schwann cells, and mesenchymal precursor cells. Macrophages at the nerve crush site are molecularly distinct from macrophages associated with Wallerian degeneration. In the injured nerve, macrophages ‘eat’ apoptotic leukocytes, a process called efferocytosis, and thereby promote an anti-inflammatory milieu. Myeloid cells in the injured nerve, but not axotomized DRGs, strongly express receptors for the cytokine GM-CSF. In GM-CSF-deficient (Csf2-/-) mice, inflammation resolution is delayed and conditioning-lesion-induced regeneration of DRG neuron central axons is abolished. Thus, carefully orchestrated inflammation resolution in the nerve is required for conditioning-lesion-induced neurorepair.
Collapse
Affiliation(s)
- Ashley L Kalinski
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Choya Yoon
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Lucas D Huffman
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States.,Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, United States
| | - Patrick C Duncker
- Department of Neurology, University of Michigan Medical School, Ann Arbor, United States
| | - Rafi Kohen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States.,Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, United States
| | - Ryan Passino
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Hannah Hafner
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Craig Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Kevin S Carbajal
- Department of Neurology, University of Michigan Medical School, Ann Arbor, United States
| | | | - Edmund Hollis
- Burke Neurological Institute, White Plains, United States.,The Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, United States.,The Neurological Institute, The Ohio State University, Columbus, United States
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, United States.,Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, United States.,Department of Neurology, University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
487
|
Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis 2020; 11:1013. [PMID: 33243969 PMCID: PMC7691519 DOI: 10.1038/s41419-020-03221-2] [Citation(s) in RCA: 617] [Impact Index Per Article: 123.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Chemotherapy, radiation therapy, as well as targeted anticancer agents can induce clinically relevant tumor-targeting immune responses, which critically rely on the antigenicity of malignant cells and their capacity to generate adjuvant signals. In particular, immunogenic cell death (ICD) is accompanied by the exposure and release of numerous damage-associated molecular patterns (DAMPs), which altogether confer a robust adjuvanticity to dying cancer cells, as they favor the recruitment and activation of antigen-presenting cells. ICD-associated DAMPs include surface-exposed calreticulin (CALR) as well as secreted ATP, annexin A1 (ANXA1), type I interferon, and high-mobility group box 1 (HMGB1). Additional hallmarks of ICD encompass the phosphorylation of eukaryotic translation initiation factor 2 subunit-α (EIF2S1, better known as eIF2α), the activation of autophagy, and a global arrest in transcription and translation. Here, we outline methodological approaches for measuring ICD markers in vitro and ex vivo for the discovery of next-generation antineoplastic agents, the development of personalized anticancer regimens, and the identification of optimal therapeutic combinations for the clinical management of cancer.
Collapse
|
488
|
Kang SG, Eskandari-Sedighi G, Hromadkova L, Safar JG, Westaway D. Cellular Biology of Tau Diversity and Pathogenic Conformers. Front Neurol 2020; 11:590199. [PMID: 33304310 PMCID: PMC7693435 DOI: 10.3389/fneur.2020.590199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022] Open
Abstract
Tau accumulation is a prominent feature in a variety of neurodegenerative disorders and remarkable effort has been expended working out the biochemistry and cell biology of this cytoplasmic protein. Tau's wayward properties may derive from germline mutations in the case of frontotemporal lobar degeneration (FTLD-MAPT) but may also be prompted by less understood cues—perhaps environmental or from molecular damage as a consequence of chronological aging—in the case of idiopathic tauopathies. Tau properties are undoubtedly affected by its covalent structure and in this respect tau protein is not only subject to changes in length produced by alternative splicing and endoproteolysis, but different types of posttranslational modifications that affect different amino acid residues. Another layer of complexity concerns alternate conformations—“conformers”—of the same covalent structures; in vivo conformers can encompass soluble oligomeric species, ramified fibrillar structures evident by light and electron microscopy and other forms of the protein that have undergone liquid-liquid phase separation to make demixed liquid droplets. Biological concepts based upon conformers have been charted previously for templated replication mechanisms for prion proteins built of the PrP polypeptide; these are now providing useful explanations to feature tau pathobiology, including how this protein accumulates within cells and how it can exhibit predictable patterns of spread across different neuroanatomical regions of an affected brain. In sum, the documented, intrinsic heterogeneity of tau forms and conformers now begins to speak to a fundamental basis for diversity in clinical presentation of tauopathy sub-types. In terms of interventions, emphasis upon subclinical events may be worthwhile, noting that irrevocable cell loss and ramified protein assemblies feature at end-stage tauopathy, whereas earlier events may offer better opportunities for diverting pathogenic processes. Nonetheless, the complexity of tau sub-types, which may be present even within intermediate disease stages, likely mitigates against one-size-fits-all therapeutic strategies and may require a suite of interventions. We consider the extent to which animal models of tauopathy can be reasonably enrolled in the campaign to produce such interventions and to slow the otherwise inexorable march of disease progression.
Collapse
Affiliation(s)
- Sang-Gyun Kang
- Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | | | - Lenka Hromadkova
- Department of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Jiri G Safar
- Department of Neurology and Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - David Westaway
- Center for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada.,Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
489
|
Snodgrass RG, Benatzy Y, Schmid T, Namgaladze D, Mainka M, Schebb NH, Lütjohann D, Brüne B. Efferocytosis potentiates the expression of arachidonate 15-lipoxygenase (ALOX15) in alternatively activated human macrophages through LXR activation. Cell Death Differ 2020; 28:1301-1316. [PMID: 33177619 PMCID: PMC8027700 DOI: 10.1038/s41418-020-00652-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022] Open
Abstract
Macrophages acquire anti-inflammatory and proresolving functions to facilitate resolution of inflammation and promote tissue repair. While alternatively activated macrophages (AAMs), also referred to as M2 macrophages, polarized by type 2 (Th2) cytokines IL-4 or IL-13 contribute to the suppression of inflammatory responses and play a pivotal role in wound healing, contemporaneous exposure to apoptotic cells (ACs) potentiates the expression of anti-inflammatory and tissue repair genes. Given that liver X receptors (LXRs), which coordinate sterol metabolism and immune cell function, play an essential role in the clearance of ACs, we investigated whether LXR activation following engulfment of ACs selectively potentiates the expression of Th2 cytokine-dependent genes in primary human AAMs. We show that AC uptake simultaneously upregulates LXR-dependent, but suppresses SREBP-2-dependent gene expression in macrophages, which are both prevented by inhibiting Niemann–Pick C1 (NPC1)-mediated sterol transport from lysosomes. Concurrently, macrophages accumulate sterol biosynthetic intermediates desmosterol, lathosterol, lanosterol, and dihydrolanosterol but not cholesterol-derived oxysterols. Using global transcriptome analysis, we identify anti-inflammatory and proresolving genes including interleukin-1 receptor antagonist (IL1RN) and arachidonate 15-lipoxygenase (ALOX15) whose expression are selectively potentiated in macrophages upon concomitant exposure to ACs or LXR agonist T0901317 (T09) and Th2 cytokines. We show priming macrophages via LXR activation enhances the cellular capacity to synthesize inflammation-suppressing specialized proresolving mediator (SPM) precursors 15-HETE and 17-HDHA as well as resolvin D5. Silencing LXRα and LXRβ in macrophages attenuates the potentiation of ALOX15 expression by concomitant stimulation of ACs or T09 and IL-13. Collectively, we identify a previously unrecognized mechanism of regulation whereby LXR integrates AC uptake to selectively shape Th2-dependent gene expression in AAMs.
Collapse
Affiliation(s)
- Ryan G Snodgrass
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Yvonne Benatzy
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Tobias Schmid
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Dmitry Namgaladze
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Malwina Mainka
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Dieter Lütjohann
- Institute for Clinical Chemistry and Clinical Pharmacology, University of Bonn, Bonn, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
490
|
Alsina-Sanchis E, Mülfarth R, Moll I, Mogler C, Rodriguez-Vita J, Fischer A. Intraperitoneal Oil Application Causes Local Inflammation with Depletion of Resident Peritoneal Macrophages. Mol Cancer Res 2020; 19:288-300. [PMID: 33139505 DOI: 10.1158/1541-7786.mcr-20-0650] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/08/2020] [Accepted: 10/27/2020] [Indexed: 11/16/2022]
Abstract
Oil is frequently used as a solvent to inject lipophilic substances into the peritoneum of laboratory animals. Although mineral oil causes chronic peritoneal inflammation, little is known whether other oils are better suited. We show that olive, peanut, corn, or mineral oil causes xanthogranulomatous inflammation with depletion of resident peritoneal macrophages. However, there were striking differences in the severity of the inflammatory response. Peanut and mineral oil caused severe chronic inflammation with persistent neutrophil and monocyte recruitment, expansion of the vasculature, and fibrosis. Corn and olive oil provoked no or only mild signs of chronic inflammation. Mechanistically, the vegetal oils were taken up by macrophages leading to foam cell formation and induction of cell death. Olive oil triggered caspase-3 cleavage and apoptosis, which facilitate the resolution of inflammation. Peanut oil and, to a lesser degree, corn oil, triggered caspase-1 activation and macrophage pyroptosis, which impair the resolution of inflammation. As such, intraperitoneal oil administration can interfere with the outcome of subsequent experiments. As a proof of principle, intraperitoneal peanut oil injection was compared with its oral delivery in a thioglycolate-induced peritonitis model. The chronic peritoneal inflammation due to peanut oil injection impeded the proper recruitment of macrophages and the resolution of inflammation in this peritonitis model. In summary, the data indicate that it is advisable to deliver lipophilic substances, like tamoxifen, by oral gavage instead of intraperitoneal injection. IMPLICATIONS: This work contributes to the reproducibility of animal research by helping to understand some of the undesired effects observed in animal experiments.
Collapse
Affiliation(s)
- Elisenda Alsina-Sanchis
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ronja Mülfarth
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Juan Rodriguez-Vita
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Andreas Fischer
- Division Vascular Signaling and Cancer (A270), German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Department of Medicine I and Clinical Chemistry, University Hospital of Heidelberg, Heidelberg, Germany.,European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
491
|
Kumar V. Understanding the complexities of SARS-CoV2 infection and its immunology: A road to immune-based therapeutics. Int Immunopharmacol 2020; 88:106980. [PMID: 33182073 PMCID: PMC7843151 DOI: 10.1016/j.intimp.2020.106980] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 02/07/2023]
Abstract
Emerging infectious diseases always pose a threat to humans along with plant and animal life. SARS-CoV2 is the recently emerged viral infection that originated from Wuhan city of the Republic of China in December 2019. Now, it has become a pandemic. Currently, SARS-CoV2 has infected more than 27.74 million people worldwide, and taken 901,928 human lives. It was named first 'WH 1 Human CoV' and later changed to 2019 novel CoV (2019-nCoV). Scientists have established it as a zoonotic viral disease emerged from Chinese horseshoe bats, which do not develop a severe infection. For example, Rhinolophus Chinese horseshoe bats harboring severe acute respiratory syndrome-related coronavirus (SARSr-CoV) or SARSr-Rh-BatCoV appear healthy and clear the virus within 2-4 months period. The article introduces first the concept of EIDs and some past EIDs, which have affected human life. Next section discusses mysteries regarding SARS-CoV2 origin, its evolution, and human transfer. Third section describes COVID-19 clinical symptoms and factors affecting susceptibility or resistance. The fourth section introduces the SARS-CoV2 entry in the host cell, its replication, and the establishment of productive infection. Section five describes the host's immune response associated with asymptomatic, symptomatic, mild to moderate, and severe COVID-19. The subsequent seventh and eighth sections mention the immune status in COVID-19 convalescent patients and re-emergence of COVID-19 in them. Thereafter, the eighth section describes viral strategies to hijack the host antiviral immune response and generate the "cytokine storm". The ninth section describes about transgenic humane ACE2 (hACE2) receptor expressing mice to study immunity, drugs, and vaccines. The article ends with the development of different immunomodulatory and immunotherapeutics strategies, including vaccines waiting for their approval in humans as prophylaxis or treatment measures.
Collapse
Affiliation(s)
- V Kumar
- Children's Health Queensland Clinical Unit, School of Clinical Medicine, Faculty of Medicine, Mater Research, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia; School of Biomedical Sciences, Faculty of Medicine, University of Queensland, ST Lucia, Brisbane, Queensland 4078, Australia.
| |
Collapse
|
492
|
Shabbir A, Rathod KS, Khambata RS, Ahluwalia A. Sex Differences in the Inflammatory Response: Pharmacological Opportunities for Therapeutics for Coronary Artery Disease. Annu Rev Pharmacol Toxicol 2020; 61:333-359. [PMID: 33035428 DOI: 10.1146/annurev-pharmtox-010919-023229] [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] [Indexed: 12/15/2022]
Abstract
Coordinated molecular responses are key to effective initiation and resolution of both acute and chronic inflammation. Vascular inflammation plays an important role in initiating and perpetuating atherosclerotic disease, specifically at the site of plaque and subsequent fibrous cap rupture. Both men and women succumb to this disease process, and although management strategies have focused on revascularization and pharmacological therapies in the acute situation to reverse vessel closure and prevent thrombogenesis, data now suggest that regulation of host inflammation may improve both morbidity and mortality, thus supporting the notion that prevention is better than cure. There is a clear sex difference in the incidence of vascular disease, and data confirm biological differences in inflammatory initiation and resolution between men and women. This article reviews contemporary opinions describing the sex difference in the initiation and resolution of inflammatory responses, with a view to explore potential targets for pharmacological intervention.
Collapse
Affiliation(s)
- Asad Shabbir
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Krishnaraj Sinhji Rathod
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Rayomand Syrus Khambata
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| | - Amrita Ahluwalia
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom;
| |
Collapse
|
493
|
Rajasinghe LD, Chauhan PS, Wierenga KA, Evered AO, Harris SN, Bates MA, Gavrilin MA, Pestka JJ. Omega-3 Docosahexaenoic Acid (DHA) Impedes Silica-Induced Macrophage Corpse Accumulation by Attenuating Cell Death and Potentiating Efferocytosis. Front Immunol 2020; 11:2179. [PMID: 33123123 PMCID: PMC7573148 DOI: 10.3389/fimmu.2020.02179] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/10/2020] [Indexed: 12/18/2022] Open
Abstract
Airway exposure of lupus-prone NZBWF1 mice to crystalline silica (cSiO2), a known trigger of human autoimmune disease, elicits sterile inflammation and alveolar macrophage death in the lung that, in turn, induces early autoimmune onset and accelerates lupus progression to fatal glomerulonephritis. Dietary supplementation with docosahexaenoic acid (DHA), a marine ω-3 polyunsaturated fatty acid (PUFA), markedly ameliorates cSiO2-triggered pulmonary, systemic, and renal manifestations of lupus. Here, we tested the hypothesis that DHA influences both cSiO2-induced death and efferocytotic clearance of resultant cell corpses using three murine macrophage models: (i) primary alveolar macrophages (AM) isolated from NZBWF1 mice; (ii) self-renewing AM-like Max Planck Institute (MPI) cells isolated from fetuses of C57BL/6 mice, and (iii) RAW 264.7 murine macrophages, a virus-transformed cell line derived from BALB/c mice stably transfected with the inflammasome adaptor protein ASC (RAW-ASC). Incubation with cSiO2 at 25 and 50 μg/ml for 6 h was found to dose-dependently induce cell death (p < 0.05) in all three models as determined by both acridine orange/propidium iodide staining and release of lactate dehydrogenase into cell culture supernatant. Pre-incubation with DHA at a physiologically relevant concentration (25 μM) significantly reduced cSiO2-induced death (p < 0.05) in all three models. Cell death induction by cSiO2 alone and its suppression by DHA were primarily associated with caspase-3/7 activation, suggestive of apoptosis, in AM, MPI, and RAW-ASC cells. Fluorescence microscopy revealed that all three macrophage models were similarly capable of efferocytosing RAW-ASC target cell corpses. Furthermore, MPI effector cells could likewise engulf RAW-ASC target cell corpses elicited by treatment with staurosporine (apoptosis), LPS, and nigericin (pyroptosis), or cSiO2. Pre-incubation of RAW-ASC target cells with 25 μM DHA prior to death induced by these agents significantly enhanced their efferocytosis (p < 0.05) by MPI effector cells. In contrast, pre-incubating MPI effector cells with DHA did not affect engulfment of RAW-ASC target cells pre-incubated with vehicle. Taken together, these findings indicate that DHA at a physiologically relevant concentration was capable of attenuating macrophage death and could potentiate efferocytosis, with the net effect of reducing accumulation of cell corpses capable of eliciting autoimmunity.
Collapse
Affiliation(s)
- Lichchavi D Rajasinghe
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Preeti S Chauhan
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Kathryn A Wierenga
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Augustus O Evered
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Shamya N Harris
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
| | - Melissa A Bates
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Mikhail A Gavrilin
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University, Columbus, OH, United States
| | - James J Pestka
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States.,Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States.,Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
494
|
Gadiyar V, Lahey KC, Calianese D, Devoe C, Mehta D, Bono K, Desind S, Davra V, Birge RB. Cell Death in the Tumor Microenvironment: Implications for Cancer Immunotherapy. Cells 2020; 9:cells9102207. [PMID: 33003477 PMCID: PMC7599747 DOI: 10.3390/cells9102207] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/26/2020] [Accepted: 09/26/2020] [Indexed: 02/06/2023] Open
Abstract
The physiological fate of cells that die by apoptosis is their prompt and efficient removal by efferocytosis. During these processes, apoptotic cells release intracellular constituents that include purine nucleotides, lysophosphatidylcholine (LPC), and Sphingosine-1-phosphate (S1P) that induce migration and chemo-attraction of phagocytes as well as mitogens and extracellular membrane-bound vesicles that contribute to apoptosis-induced compensatory proliferation and alteration of the extracellular matrix and the vascular network. Additionally, during efferocytosis, phagocytic cells produce a number of anti-inflammatory and resolving factors, and, together with apoptotic cells, efferocytic events have a homeostatic function that regulates tissue repair. These homeostatic functions are dysregulated in cancers, where, aforementioned events, if not properly controlled, can lead to cancer progression and immune escape. Here, we summarize evidence that apoptosis and efferocytosis are exploited in cancer, as well as discuss current translation and clinical efforts to harness signals from dying cells into therapeutic strategies.
Collapse
|
495
|
Wang L, Li H, Tang Y, Yao P. Potential Mechanisms and Effects of Efferocytosis in Atherosclerosis. Front Endocrinol (Lausanne) 2020; 11:585285. [PMID: 33597922 PMCID: PMC7883484 DOI: 10.3389/fendo.2020.585285] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/14/2020] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis (AS) is the main pathological basis for the development of cardio-cerebrovascular diseases. Abnormal accumulation of apoptotic and necrotic cells resulted in plaque enlargement, necrotic core formation and plaque rupture in AS. Under physiological conditions, apoptotic cells (ACs) could be effectively phagocytized and cleared by phagocyte-mediated efferocytosis. In contrast, the clearance efficiency of ACs in AS plaque was much lower because of the impaired efferocytosis in AS. Recent findings have made great progress on the molecular mechanisms of efferocytosis process and dynamic regulation, and its dysfunction on organismal health. Yet, there are still few effective treatments for this process. This article reviews the mechanism of efferocytosis and the role of efferocytosis in AS, highlighting a novel therapeutic strategy for AS, which mainly prevents the progression of plaque by targeting efferocytosis.
Collapse
Affiliation(s)
- Lili Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongxia Li
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuhan Tang
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yao
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Ministry of Education Key Laboratory of Environment, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
496
|
Green DR. Dances with Cells. Crit Rev Immunol 2020; 40:355-366. [PMID: 33463948 PMCID: PMC11019855 DOI: 10.1615/critrevimmunol.2020035038] [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] [Indexed: 11/13/2022]
Abstract
Like all of us, cells proceed through stages of life. In this whimsical review, aspects of several such stages, including birth, growth, aging, death, and "afterlife," are considered, with a special emphasis on cells of the immune system. Discussed along the way are asymmetric division of activated, naive cluster of differentiation 8+ T cells, c-Myc and polyamines in lymphocyte function and aging, cell survival after induction of cell death pathways, cell death consequences, and clearance of dead cells from surrounding tissues. This is offered in memory of the unique and wonderful Dr. Eli Sercarz.
Collapse
Affiliation(s)
- Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| |
Collapse
|