1
|
Chen X, Zhang L, Chai W, Tian P, Kim J, Ding J, Zhang H, Liu C, Wang D, Cui X, Pan H. Hypoxic Microenvironment Reconstruction with Synergistic Biofunctional Ions Promotes Diabetic Wound Healing. Adv Healthc Mater 2023; 12:e2301984. [PMID: 37740829 DOI: 10.1002/adhm.202301984] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/14/2023] [Indexed: 09/25/2023]
Abstract
Chronic hypoxia and ischemia make diabetic wounds non-healing. Cellular functions of diabetic chronic wounds are inhibited under a pathological environment. Therefore, this work develops a composite hydrogel system to promote diabetic wound healing. The composite hydrogel system consists of ε-poly-lysine (EPL), calcium peroxide (CP), and borosilicate glass (BG). The hydrogel supplies continuous dissolved oxygen molecules to the wound that can penetrate the skin tissue to restore normal cellular function and promote vascular regeneration. Biofunctional ions released from BGs can recruit more macrophages through neovascularization and modulate macrophage phenotypic transformation. Combining oxygen-mediated vascular regeneration and ion-mediated inflammatory regulation significantly accelerated diabetic wound healing. These findings indicate that this composite hydrogel system holds promise as a novel tissue engineering material.
Collapse
Affiliation(s)
- Xiaochen Chen
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Liyan Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Wenwen Chai
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Pengfei Tian
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jua Kim
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Jingxin Ding
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Hao Zhang
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Chunyu Liu
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Deping Wang
- School of materials science and engineering, Tongji University, Shanghai, 201804, P. R. China
| | - Xu Cui
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Haobo Pan
- Center for Human Tissues and Organs Degeneration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- Shenzhen Healthemes Biotechnology Co., Ltd, Shenzhen, 518071, P. R. China
| |
Collapse
|
2
|
Oates TCL, Moura PL, Cross S, Roberts K, Baum HE, Haydn‐Smith KL, Wilson MC, Heesom KJ, Severn CE, Toye AM. Defining the proteomic landscape of cultured macrophages and their polarization continuum. Immunol Cell Biol 2023; 101:947-963. [PMID: 37694300 PMCID: PMC10953363 DOI: 10.1111/imcb.12687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/28/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023]
Abstract
Macrophages have previously been characterized based on phenotypical and functional differences into suggested simplified subtypes of MØ, M1, M2a and M2c. These macrophage subtypes can be generated in a well-established primary monocyte culture model that produces cells expressing accepted subtype surface markers. To determine how these subtypes retain functional similarities and better understand their formation, we generated all four subtypes from the same donors. Comparative whole-cell proteomics confirmed that four distinct macrophage subtypes could be induced from the same donor material, with > 50% of 5435 identified proteins being significantly altered in abundance between subtypes. Functional assessment highlighted that these distinct protein expression profiles are primed to enable specific cell functions, indicating that this shifting proteome is predictive of meaningful changes in cell characteristics. Importantly, the 2552 proteins remained consistent in abundance across all macrophage subtypes examined, demonstrating maintenance of a stable core proteome that likely enables swift polarity changes. We next explored the cross-polarization capabilities of preactivated M1 macrophages treated with dexamethasone. Importantly, these treated cells undergo a partial repolarization toward the M2c surface markers but still retain the M1 functional phenotype. Our investigation of polarized macrophage subtypes therefore provides evidence of a sliding scale of macrophage functionality, with these data sets providing a valuable benchmark resource for further studies of macrophage polarity, with relevance for cell therapy development and drug discovery.
Collapse
Affiliation(s)
- Tiah CL Oates
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| | - Pedro L Moura
- Center for Haematology and Regenerative Medicine, Department of Medicine (MedH)Karolinska InstitutetHuddingeSweden
| | | | - Kiren Roberts
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - Holly E Baum
- Max Planck Bristol Centre for Minimal Biology, School of ChemistryUniversity of BristolBristolUK
| | - Katy L Haydn‐Smith
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | | | - Kate J Heesom
- Proteomics Facility, Biomedical Sciences BuildingUniversity of BristolBristolUK
| | - Charlotte E Severn
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences BuildingUniversity of BristolBristolUK
- National Institute for Health Research Blood and Transplant Research Unit (NIHR BTRU) in Red Blood Cell ProductsUniversity of BristolBristolUK
| |
Collapse
|
3
|
Ryan EM, Sadiku P, Coelho P, Watts ER, Zhang A, Howden AJM, Sanchez-Garcia MA, Bewley M, Cole J, McHugh BJ, Vermaelen W, Ghesquiere B, Carmeliet P, Rodriguez Blanco G, Von Kriegsheim A, Sanchez Y, Rumsey W, Callahan JF, Cooper G, Parkinson N, Baillie K, Cantrell DA, McCafferty J, Choudhury G, Singh D, Dockrell DH, Whyte MKB, Walmsley SR. NRF2 Activation Reprograms Defects in Oxidative Metabolism to Restore Macrophage Function in Chronic Obstructive Pulmonary Disease. Am J Respir Crit Care Med 2023; 207:998-1011. [PMID: 36724365 PMCID: PMC7614437 DOI: 10.1164/rccm.202203-0482oc] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Rationale: Chronic obstructive pulmonary disease (COPD) is a disease characterized by persistent airway inflammation and disordered macrophage function. The extent to which alterations in macrophage bioenergetics contribute to impaired antioxidant responses and disease pathogenesis has yet to be fully delineated. Objectives: Through the study of COPD alveolar macrophages (AMs) and peripheral monocyte-derived macrophages (MDMs), we sought to establish if intrinsic defects in core metabolic processes drive macrophage dysfunction and redox imbalance. Methods: AMs and MDMs from donors with COPD and healthy donors underwent functional, metabolic, and transcriptional profiling. Measurements and Main Results: We observed that AMs and MDMs from donors with COPD display a critical depletion in glycolytic- and mitochondrial respiration-derived energy reserves and an overreliance on glycolysis as a source for ATP, resulting in reduced energy status. Defects in oxidative metabolism extend to an impaired redox balance associated with defective expression of the NADPH-generating enzyme, ME1 (malic enzyme 1), a known target of the antioxidant transcription factor NRF2 (nuclear factor erythroid 2-related factor 2). Consequently, selective activation of NRF2 resets the COPD transcriptome, resulting in increased generation of TCA cycle intermediaries, improved energetic status, favorable redox balance, and recovery of macrophage function. Conclusions: In COPD, an inherent loss of metabolic plasticity leads to metabolic exhaustion and reduced redox capacity, which can be rescued by activation of the NRF2 pathway. Targeting these defects, via NRF2 augmentation, may therefore present an attractive therapeutic strategy for the treatment of the aberrant airway inflammation described in COPD.
Collapse
Affiliation(s)
- Eilise M. Ryan
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Pranvera Sadiku
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Patricia Coelho
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Emily R. Watts
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Ailiang Zhang
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Andrew J. M. Howden
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, United Kingdom
| | - Manuel A. Sanchez-Garcia
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Martin Bewley
- Department of Infection, Immunity, and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Joby Cole
- Department of Infection, Immunity, and Cardiovascular Disease, University of Sheffield, Sheffield, United Kingdom
| | - Brian J. McHugh
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Wesley Vermaelen
- Metabolomics Expertise Centre, VIB-KU Leuven Centre for Cancer Biology, Leuven, Belgium
| | - Bart Ghesquiere
- Metabolomics Expertise Centre, VIB-KU Leuven Centre for Cancer Biology, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Centre for Cancer Biology, VIB, Department of Oncology, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | | | | | - Yolanda Sanchez
- GlaxoSmithKline Research & Development, Collegeville, Pennsylvania
| | - William Rumsey
- GlaxoSmithKline Research & Development, Collegeville, Pennsylvania
| | | | - George Cooper
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Nicholas Parkinson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Kenneth Baillie
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Doreen A. Cantrell
- Division of Cell Signalling and Immunology, University of Dundee, Dundee, United Kingdom
| | - John McCafferty
- NHS Lothian, Respiratory Medicine, Edinburgh, United Kingdom; and
| | - Gourab Choudhury
- NHS Lothian, Respiratory Medicine, Edinburgh, United Kingdom; and
| | - Dave Singh
- Division of Infection, Immunity, and Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - David H. Dockrell
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Moira K. B. Whyte
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| | - Sarah R. Walmsley
- University of Edinburgh Centre for Inflammation Research, The Queen’s Medical Research Institute
| |
Collapse
|
4
|
Saas P, Fan GC. Editorial: Hypoxia and inflammation: A two-way street. Front Immunol 2023; 14:1171116. [PMID: 36969222 PMCID: PMC10031103 DOI: 10.3389/fimmu.2023.1171116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/01/2023] [Indexed: 03/10/2023] Open
Affiliation(s)
- Philippe Saas
- EFS, Recherche et Développement, Grenoble, France
- Université Grenoble-Alpes, INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
- *Correspondence: Philippe Saas, ; Guo-Chang Fan,
| | - Guo-Chang Fan
- Department of Pharmacology & Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
- *Correspondence: Philippe Saas, ; Guo-Chang Fan,
| |
Collapse
|
5
|
Macrophage Phenotyping in Atherosclerosis by Proteomics. Int J Mol Sci 2023; 24:ijms24032613. [PMID: 36768933 PMCID: PMC9917096 DOI: 10.3390/ijms24032613] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Macrophages are heterogeneous and plastic cells, able to adapt their phenotype and functions to changes in the microenvironment. They are involved in several homeostatic processes and also in many human diseases, including atherosclerosis, where they participate in all the stages of the disease. For these reasons, macrophages have been studied extensively using different approaches, including proteomics. Proteomics, indeed, may be a powerful tool to better understand the behavior of these cells, and a careful analysis of the proteome of different macrophage phenotypes can help to better characterize the role of these phenotypes in atherosclerosis and provide a broad view of proteins that might potentially affect the course of the disease. In this review, we discuss the different proteomic techniques that have been used to delineate the proteomic profile of macrophage phenotypes and summarize some results that can help to elucidate the roles of macrophages and develop new strategies to counteract the progression of atherosclerosis and/or promote regression.
Collapse
|
6
|
Brace N, Megson IL, Rossi AG, Doherty MK, Whitfield PD. SILAC-based quantitative proteomics to investigate the eicosanoid associated inflammatory response in activated macrophages. J Inflamm (Lond) 2022; 19:12. [PMID: 36050729 PMCID: PMC9438320 DOI: 10.1186/s12950-022-00309-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/10/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Macrophages play a central role in inflammation by phagocytosing invading pathogens, apoptotic cells and debris, as well as mediating repair of tissues damaged by trauma. In order to do this, these dynamic cells generate a variety of inflammatory mediators including eicosanoids such as prostaglandins, leukotrienes and hydroxyeicosatraenoic acids (HETEs) that are formed through the cyclooxygenase, lipoxygenase and cytochrome P450 pathways. The ability to examine the effects of eicosanoid production at the protein level is therefore critical to understanding the mechanisms associated with macrophage activation. RESULTS This study presents a stable isotope labelling with amino acids in cell culture (SILAC) -based proteomics strategy to quantify the changes in macrophage protein abundance following inflammatory stimulation with Kdo2-lipid A and ATP, with a focus on eicosanoid metabolism and regulation. Detailed gene ontology analysis, at the protein level, revealed several key pathways with a decrease in expression in response to macrophage activation, which included a promotion of macrophage polarisation and dynamic changes to energy requirements, transcription and translation. These findings suggest that, whilst there is evidence for the induction of a pro-inflammatory response in the form of prostaglandin secretion, there is also metabolic reprogramming along with a change in cell polarisation towards a reduced pro-inflammatory phenotype. CONCLUSIONS Advanced quantitative proteomics in conjunction with functional pathway network analysis is a useful tool to investigate the molecular pathways involved in inflammation.
Collapse
Affiliation(s)
- Nicole Brace
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Ian L Megson
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Adriano G Rossi
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Mary K Doherty
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK
| | - Phillip D Whitfield
- Division of Biomedical Sciences, University of the Highlands and Islands, Centre for Health Science, Old Perth Road, Inverness, IV2 3JH, UK.
- Present Address: Glasgow Polyomics, Garscube Campus, University of Glasgow, Glasgow, G61 1BD, UK.
| |
Collapse
|
7
|
Volk RF, Montaño JL, Warrington SE, Hofmann KL, Zaro BW. Proteomic characterization of phagocytic primary human monocyte-derived macrophages. RSC Chem Biol 2022; 3:783-793. [PMID: 35755185 PMCID: PMC9175098 DOI: 10.1039/d2cb00076h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/12/2022] [Indexed: 11/21/2022] Open
Abstract
Macrophages play a vital role in the innate immune system, identifying and destroying unwanted cells. However, it has been difficult to attain a comprehensive understanding of macrophage protein abundance due to technical limitations. In addition, it remains unclear how changes in proteome composition are linked to phagocytic activity. In this study we developed methods to derive human macrophages and prepare them for mass spectrometry analysis in order to more-deeply understand the proteomic consequences of macrophage stimulation. Interferon gamma (IF-g), an immune stimulating cytokine, was used to induce macrophage activation, increasing phagocytosis of cancer cells by 2-fold. These conditions were used to perform comparative shotgun proteomics between resting macrophages and stimulated macrophages with increased phagocytic activity. Our analysis revealed that macrophages bias their protein production toward biological processes associated with phagocytosis and antigen processing in response to stimulation. We confirmed our findings by antibody-based western blotting experiments, validating both previously reported and novel proteins of interest. In addition to whole protein changes, we evaluated active protein synthesis by treating cells with the methionine surrogate probe homopropargylglycine (HPG). We saw increased rates of HPG incorporation during phagocytosis-inducing stimulation, suggesting protein synthesis rates are altered by stimulation. Together our findings provide the most comprehensive proteomic insight to date into primary human macrophages. We anticipate that this data can be used as a launchpoint to generate new hypotheses about innate immune function.
Collapse
Affiliation(s)
- Regan F Volk
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| | - José L Montaño
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| | - Sara E Warrington
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| | - Katherine L Hofmann
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| | - Balyn W Zaro
- Department of Pharmaceutical Chemistry, Cardiovascular Research Institute, Quantitative Biosciences Institute, and Helen Diller Family Comprehensive Cancer Centre, University of California San Francisco CA USA
| |
Collapse
|
8
|
Malier M, Gharzeddine K, Laverriere MH, Decaens T, Roth G, Millet A. [Tumor-associated macrophages: New targets to thwart 5-FU chemoresistance in colorectal cancers?]. Med Sci (Paris) 2022; 38:243-245. [PMID: 35333158 DOI: 10.1051/medsci/2022017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Marie Malier
- Université Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Institut pour l'avancée des biosciences, Boulevard de la Chantourne, 38700 La Tronche, France - Équipe de mécanobiologie, immunité et cancer, Institut pour l'avancée des biosciences
| | - Khaldoun Gharzeddine
- Université Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Institut pour l'avancée des biosciences, Boulevard de la Chantourne, 38700 La Tronche, France - Équipe de mécanobiologie, immunité et cancer, Institut pour l'avancée des biosciences
| | - Marie-Hélène Laverriere
- Université Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Institut pour l'avancée des biosciences, Boulevard de la Chantourne, 38700 La Tronche, France - Équipe de mécanobiologie, immunité et cancer, Institut pour l'avancée des biosciences - Département d'analyse cytologique et pathologique, CHU Grenoble Alpes, Grenoble, France
| | - Thomas Decaens
- Université Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Institut pour l'avancée des biosciences, Boulevard de la Chantourne, 38700 La Tronche, France - Service d'hépato-gastro-entérologie, CHU Grenoble Alpes, Grenoble, France
| | - Gael Roth
- Université Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Institut pour l'avancée des biosciences, Boulevard de la Chantourne, 38700 La Tronche, France - Équipe de mécanobiologie, immunité et cancer, Institut pour l'avancée des biosciences - Service d'hépato-gastro-entérologie, CHU Grenoble Alpes, Grenoble, France
| | - Arnaud Millet
- Université Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Institut pour l'avancée des biosciences, Boulevard de la Chantourne, 38700 La Tronche, France - Équipe de mécanobiologie, immunité et cancer, Institut pour l'avancée des biosciences - Service d'hépato-gastro-entérologie, CHU Grenoble Alpes, Grenoble, France
| |
Collapse
|
9
|
Eades L, Drozd M, Cubbon RM. Hypoxia signalling in the regulation of innate immune training. Biochem Soc Trans 2022; 50:413-422. [PMID: 35015075 PMCID: PMC9022967 DOI: 10.1042/bst20210857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022]
Abstract
Innate immune function is shaped by prior exposures in a phenomenon often referred to as 'memory' or 'training'. Diverse stimuli, ranging from pathogen-associated molecules to atherogenic lipoproteins, induce long-lasting training, impacting on future responses, even to distinct stimuli. It is now recognised that epigenetic modifications in innate immune cells, and their progenitors, underpin these sustained behavioural changes, and that rewired cellular metabolism plays a key role in facilitating such epigenetic marks. Oxygen is central to cellular metabolism, and cells exposed to hypoxia undergo profound metabolic rewiring. A central effector of these responses are the hypoxia inducible factors (or HIFs), which drive transcriptional programmes aiming to adapt cellular homeostasis, such as by increasing glycolysis. These metabolic shifts indirectly promote post-translational modification of the DNA-binding histone proteins, and also of DNA itself, which are retained even after cellular oxygen tension and metabolism normalise, chronically altering DNA accessibility and utilisation. Notably, the activity of HIFs can be induced in some normoxic circumstances, indicating their broad importance to cell biology, irrespective of oxygen tension. Some HIFs are implicated in innate immune training and hypoxia is present in many disease states, yet many questions remain about the association between hypoxia and training, both in health and disease. Moreover, it is now appreciated that cellular responses to hypoxia are mediated by non-HIF pathways, suggesting that other mechanisms of training may be possible. This review sets out to define what is already known about the topic, address gaps in our knowledge, and provide recommendations for future research.
Collapse
Affiliation(s)
- Lauren Eades
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Clarendon Way, Leeds LS2 9JT, U.K
| | - Michael Drozd
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Clarendon Way, Leeds LS2 9JT, U.K
| | - Richard M. Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, The University of Leeds, Clarendon Way, Leeds LS2 9JT, U.K
| |
Collapse
|
10
|
Nguyen TN, Siddiqui G, Veldhuis NA, Poole DP. Diverse Roles of TRPV4 in Macrophages: A Need for Unbiased Profiling. Front Immunol 2022; 12:828115. [PMID: 35126384 PMCID: PMC8811046 DOI: 10.3389/fimmu.2021.828115] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 12/24/2021] [Indexed: 12/27/2022] Open
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a non-selective mechanosensitive ion channel expressed by various macrophage populations. Recent reports have characterized the role of TRPV4 in shaping the activity and phenotype of macrophages to influence the innate immune response to pathogen exposure and inflammation. TRPV4 has been studied extensively in the context of inflammation and inflammatory pain. Although TRPV4 activity has been generally described as pro-inflammatory, emerging evidence suggests a more complex role where this channel may also contribute to anti-inflammatory activities. However, detailed understanding of how TRPV4 may influence the initiation, maintenance, and resolution of inflammatory disease remains limited. This review highlights recent insights into the cellular processes through which TRPV4 contributes to pathological conditions and immune processes, with a focus on macrophage biology. The potential use of high-throughput and omics methods as an unbiased approach for studying the functional outcomes of TRPV4 activation is also discussed.
Collapse
Affiliation(s)
- Thanh-Nhan Nguyen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Nicholas A. Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- *Correspondence: Daniel P. Poole, ; Nicholas A. Veldhuis,
| | - Daniel P. Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Australian Research Council (ARC) Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, VIC, Australia
- *Correspondence: Daniel P. Poole, ; Nicholas A. Veldhuis,
| |
Collapse
|
11
|
Malier M, Gharzeddine K, Laverriere MH, Marsili S, Thomas F, Decaens T, Roth G, Millet A. Hypoxia Drives Dihydropyrimidine Dehydrogenase Expression in Macrophages and Confers Chemoresistance in Colorectal Cancer. Cancer Res 2021; 81:5963-5976. [PMID: 34645611 PMCID: PMC9397622 DOI: 10.1158/0008-5472.can-21-1572] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/17/2021] [Accepted: 10/07/2021] [Indexed: 01/07/2023]
Abstract
Colorectal adenocarcinoma is a leading cause of death worldwide, and immune infiltration in colorectal tumors has been recognized recently as an important pathophysiologic event. In this context, tumor-associated macrophages (TAM) have been related to chemoresistance to 5-fluorouracil (5-FU), the first-line chemotherapeutic agent used in treating colorectal cancers. Nevertheless, the details of this chemoresistance mechanism are still poorly elucidated. In the current study, we report that macrophages specifically overexpress dihydropyrimidine dehydrogenase (DPD) in hypoxia, leading to macrophage-induced chemoresistance to 5-FU via inactivation of the drug. Hypoxia-induced macrophage DPD expression was controlled by HIF2α. TAMs constituted the main contributors to DPD activity in human colorectal primary or secondary tumors, while cancer cells did not express significant levels of DPD. In addition, contrary to humans, macrophages in mice do not express DPD. Together, these findings shed light on the role of TAMs in promoting chemoresistance in colorectal cancers and identify potential new therapeutic targets. SIGNIFICANCE: Hypoxia induces HIF2α-mediated overexpression of dihydropyrimidine dehydrogenase in TAMs, leading to chemoresistance to 5-FU in colon cancers.
Collapse
Affiliation(s)
- Marie Malier
- Team Mechanobiology, Immunity and Cancer, Institute for Advanced Biosciences Inserm 1209 – UMR CNRS 5309, Grenoble, France.,Grenoble Alpes University, Grenoble, France.,Department of Hepatogastroenterology, University Hospital Grenoble-Alpes, Grenoble, France
| | - Khaldoun Gharzeddine
- Team Mechanobiology, Immunity and Cancer, Institute for Advanced Biosciences Inserm 1209 – UMR CNRS 5309, Grenoble, France.,Grenoble Alpes University, Grenoble, France.,Research Department, University Hospital Grenoble-Alpes, Grenoble, France
| | - Marie-Hélène Laverriere
- Team Mechanobiology, Immunity and Cancer, Institute for Advanced Biosciences Inserm 1209 – UMR CNRS 5309, Grenoble, France.,Grenoble Alpes University, Grenoble, France.,Department of Pathological Anatomy and Cytology, University Hospital Grenoble-Alpes, Grenoble, France
| | - Sabrina Marsili
- CRCT Inserm U037, Toulouse University 3, Toulouse, France.,Claudius Regaud Institute, IUCT-Oncopole, Toulouse, France
| | - Fabienne Thomas
- CRCT Inserm U037, Toulouse University 3, Toulouse, France.,Claudius Regaud Institute, IUCT-Oncopole, Toulouse, France
| | - Thomas Decaens
- Grenoble Alpes University, Grenoble, France.,Department of Hepatogastroenterology, University Hospital Grenoble-Alpes, Grenoble, France.,Team Tumor Molecular Pathology and Biomarkers, Institute for Advanced Biosciences UMR Inserm 1209 – CNRS 5309, Grenoble, France
| | - Gael Roth
- Team Mechanobiology, Immunity and Cancer, Institute for Advanced Biosciences Inserm 1209 – UMR CNRS 5309, Grenoble, France.,Grenoble Alpes University, Grenoble, France.,Department of Hepatogastroenterology, University Hospital Grenoble-Alpes, Grenoble, France
| | - Arnaud Millet
- Team Mechanobiology, Immunity and Cancer, Institute for Advanced Biosciences Inserm 1209 – UMR CNRS 5309, Grenoble, France.,Grenoble Alpes University, Grenoble, France.,Department of Hepatogastroenterology, University Hospital Grenoble-Alpes, Grenoble, France.,Corresponding Author: Arnaud Millet, Institute for Advanced Biosciences, Grenoble 38000, France. Phone: 33-6-66-88-34-82; E-mail:
| |
Collapse
|
12
|
He L, Jhong JH, Chen Q, Huang KY, Strittmatter K, Kreuzer J, DeRan M, Wu X, Lee TY, Slavov N, Haas W, Marneros AG. Global characterization of macrophage polarization mechanisms and identification of M2-type polarization inhibitors. Cell Rep 2021; 37:109955. [PMID: 34731634 PMCID: PMC8783961 DOI: 10.1016/j.celrep.2021.109955] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 08/20/2021] [Accepted: 10/15/2021] [Indexed: 01/07/2023] Open
Abstract
Macrophages undergoing M1- versus M2-type polarization differ significantly in their cell metabolism and cellular functions. Here, global quantitative time-course proteomics and phosphoproteomics paired with transcriptomics provide a comprehensive characterization of temporal changes in cell metabolism, cellular functions, and signaling pathways that occur during the induction phase of M1- versus M2-type polarization. Significant differences in, especially, metabolic pathways are observed, including changes in glucose metabolism, glycosaminoglycan metabolism, and retinoic acid signaling. Kinase-enrichment analysis shows activation patterns of specific kinases that are distinct in M1- versus M2-type polarization. M2-type polarization inhibitor drug screens identify drugs that selectively block M2- but not M1-type polarization, including mitogen-activated protein kinase kinase (MEK) and histone deacetylase (HDAC) inhibitors. These datasets provide a comprehensive resource to identify specific signaling and metabolic pathways that are critical for macrophage polarization. In a proof-of-principle approach, we use these datasets to show that MEK signaling is required for M2-type polarization by promoting peroxisome proliferator-activated receptor-γ (PPARγ)-induced retinoic acid signaling.
Collapse
Affiliation(s)
- Lizhi He
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jhih-Hua Jhong
- Department of Computer Science and Engineering, Yuan Ze University, Taoyuan 320, Taiwan; Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Qi Chen
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Kai-Yao Huang
- Department of Medical Research, Hsinchu Mackay Memorial Hospital, Hsinchu 300, Taiwan
| | - Karin Strittmatter
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Johannes Kreuzer
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Michael DeRan
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Xu Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA
| | - Tzong-Yi Lee
- Warshel Institute for Computational Biology, School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen 518172, China
| | - Nikolai Slavov
- Department of Bioengineering and Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Wilhelm Haas
- Cancer Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | - Alexander G Marneros
- Cutaneous Biology Research Center, Massachusetts General Hospital, and Department of Dermatology, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
13
|
Molendijk J, Seldin MM, Parker BL. CoffeeProt: an online tool for correlation and functional enrichment of systems genetics data. Nucleic Acids Res 2021; 49:W104-W113. [PMID: 33978718 PMCID: PMC8262721 DOI: 10.1093/nar/gkab352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/08/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
The integration of genomics, transcriptomics, proteomics and phenotypic traits across genetically diverse populations is a powerful approach to discover novel biological regulators. The increasing volume of complex data require new and easy-to-use tools accessible to a variety of scientists for the discovery and visualization of functionally relevant associations. To meet this requirement, we developed CoffeeProt, an open-source tool that analyses genetic variants associated to protein networks, other omics datatypes and phenotypic traits. CoffeeProt uses transcriptomics or proteomics data to perform correlation network analyses and annotates results with protein-protein interactions, subcellular localisations and drug associations. It then integrates genetic variants associated with gene expression (eQTLs) or protein abundance (pQTLs) and includes predictions of the potential consequences of variants on gene function. Finally, genetic variants are co-mapped to molecular or phenotypic traits either provided by the user or retrieved directly from publicly available GWAS results. We demonstrate its utility with the analysis of mouse and human population data enabling the rapid identification of genetic variants associated with druggable proteins and clinical traits. We expect that CoffeeProt will serve the systems genetics and basic science research communities, leading to the discovery of novel biologically relevant associations. CoffeeProt is available at www.coffeeprot.com.
Collapse
Affiliation(s)
- Jeffrey Molendijk
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Marcus M Seldin
- Department of Biological Chemistry and Center for Epigenetics and Metabolism, University of California, Irvine, CA 92697, USA
| | - Benjamin L Parker
- Department of Anatomy and Physiology, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
14
|
Pinto SM, Kim H, Subbannayya Y, Giambelluca MS, Bösl K, Ryan L, Sharma A, Kandasamy RK. Comparative Proteomic Analysis Reveals Varying Impact on Immune Responses in Phorbol 12-Myristate-13-Acetate-Mediated THP-1 Monocyte-to-Macrophage Differentiation. Front Immunol 2021; 12:679458. [PMID: 34234780 PMCID: PMC8255674 DOI: 10.3389/fimmu.2021.679458] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/25/2021] [Indexed: 01/06/2023] Open
Abstract
Macrophages are sentinels of the innate immune system, and the human monocytic cell line THP-1 is one of the widely used in vitro models to study inflammatory processes and immune responses. Several monocyte-to-macrophage differentiation protocols exist, with phorbol 12-myristate-13-acetate (PMA) being the most commonly used and accepted method. However, the concentrations and duration of PMA treatment vary widely in the published literature and could affect the probed phenotype, however their effect on protein expression is not fully deciphered. In this study, we employed a dimethyl labeling-based quantitative proteomics approach to determine the changes in the protein repertoire of macrophage-like cells differentiated from THP-1 monocytes by three commonly used PMA-based differentiation protocols. Employing an integrated network analysis, we show that variations in PMA concentration and duration of rest post-stimulation result in downstream differences in the protein expression and cellular signaling processes. We demonstrate that these differences result in altered inflammatory responses, including variation in the expression of cytokines upon stimulation with various Toll-like receptor (TLR) agonists. Together, these findings provide a valuable resource that significantly expands the knowledge of protein expression dynamics with one of the most common in vitro models for macrophages, which in turn has a profound impact on the immune as well as inflammatory responses being studied.
Collapse
Affiliation(s)
- Sneha M. Pinto
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - Hera Kim
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Yashwanth Subbannayya
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Miriam S. Giambelluca
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Korbinian Bösl
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
- Department of Infectious Diseases, Medical Clinic, St. Olavs Hospital, Trondheim, Norway
| | - Liv Ryan
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Animesh Sharma
- Proteomics and Modomics Experimental Core, PROMEC, Norwegian University of Science and Technology and the Central Norway Regional Health Authority, Stjørdal, Norway
| | - Richard K. Kandasamy
- Centre of Molecular Inflammation Research (CEMIR), and Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
15
|
Urrutia PJ, Bórquez DA, Núñez MT. Inflaming the Brain with Iron. Antioxidants (Basel) 2021; 10:antiox10010061. [PMID: 33419006 PMCID: PMC7825317 DOI: 10.3390/antiox10010061] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 02/06/2023] Open
Abstract
Iron accumulation and neuroinflammation are pathological conditions found in several neurodegenerative diseases, including Alzheimer's disease (AD) and Parkinson's disease (PD). Iron and inflammation are intertwined in a bidirectional relationship, where iron modifies the inflammatory phenotype of microglia and infiltrating macrophages, and in turn, these cells secrete diffusible mediators that reshape neuronal iron homeostasis and regulate iron entry into the brain. Secreted inflammatory mediators include cytokines and reactive oxygen/nitrogen species (ROS/RNS), notably hepcidin and nitric oxide (·NO). Hepcidin is a small cationic peptide with a central role in regulating systemic iron homeostasis. Also present in the cerebrospinal fluid (CSF), hepcidin can reduce iron export from neurons and decreases iron entry through the blood-brain barrier (BBB) by binding to the iron exporter ferroportin 1 (Fpn1). Likewise, ·NO selectively converts cytosolic aconitase (c-aconitase) into the iron regulatory protein 1 (IRP1), which regulates cellular iron homeostasis through its binding to iron response elements (IRE) located in the mRNAs of iron-related proteins. Nitric oxide-activated IRP1 can impair cellular iron homeostasis during neuroinflammation, triggering iron accumulation, especially in the mitochondria, leading to neuronal death. In this review, we will summarize findings that connect neuroinflammation and iron accumulation, which support their causal association in the neurodegenerative processes observed in AD and PD.
Collapse
Affiliation(s)
- Pamela J. Urrutia
- Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024 Santiago, Chile;
| | - Daniel A. Bórquez
- Center for Biomedical Research, Faculty of Medicine, Universidad Diego Portales, 8370007 Santiago, Chile;
| | - Marco Tulio Núñez
- Department of Biology, Faculty of Sciences, Universidad de Chile, 7800024 Santiago, Chile;
- Correspondence: ; Tel.: +56-2-29787360
| |
Collapse
|
16
|
Ahmed I, Ismail N. M1 and M2 Macrophages Polarization via mTORC1 Influences Innate Immunity and Outcome of Ehrlichia Infection. JOURNAL OF CELLULAR IMMUNOLOGY 2020; 2:108-115. [PMID: 32719831 DOI: 10.33696/immunology.2.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Human monocytic ehrlichiosis (HME) is an emerging life-threatening tick-borne disease caused by the obligate intracellular bacterium Ehrlichia chaffeensis. HME is often presented as a nonspecific flu-like illness characterized by presence of fever, headache, malaise, and myalgia. However, in some cases the disease can evolve to a severe form, which is commonly marked by acute liver injury followed by multi-organ failure and toxic shock-like syndrome [1-3]. Macrophages and monocytes are the major target cells for Ehrlichia, although this bacterium can infect other cell types such as hepatocytes and endothelial cells [4]. In this article, we discuss how macrophages polarization to M1 or M2 phenotypes dictate the severity of ehrlichiosis and the outcome of infection. We will also discuss the potential mechanisms that regulate such polarization.
Collapse
Affiliation(s)
- Ibrahim Ahmed
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Nahed Ismail
- Department of Pathology, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
17
|
Hinneburg H, Pedersen JL, Bokil NJ, Pralow A, Schirmeister F, Kawahara R, Rapp E, Saunders BM, Thaysen-Andersen M. High-resolution longitudinal N- and O-glycoprofiling of human monocyte-to-macrophage transition. Glycobiology 2020; 30:679-694. [DOI: 10.1093/glycob/cwaa020] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Protein glycosylation impacts the development and function of innate immune cells. The glycophenotypes and the glycan remodelling associated with the maturation of macrophages from monocytic precursor populations remain incompletely described. Herein, label-free porous graphitised carbon–liquid chromatography–tandem mass spectrometry (PGC-LC-MS/MS) was employed to profile with high resolution the N- and O-glycome associated with human monocyte-to-macrophage transition. Primary blood-derived CD14+ monocytes were differentiated ex vivo in the absence of strong anti- and proinflammatory stimuli using a conventional 7-day granulocyte-macrophage colony-stimulating factor differentiation protocol with longitudinal sampling. Morphology and protein expression monitored by light microscopy and proteomics validated the maturation process. Glycomics demonstrated that monocytes and macrophages display similar N-glycome profiles, comprising predominantly paucimannosidic (Man1-3GlcNAc2Fuc0–1, 22.1–30.8%), oligomannosidic (Man5-9GlcNAc2, 29.8–35.7%) and α2,3/6-sialylated complex-type N-glycans with variable core fucosylation (27.6–39.1%). Glycopeptide analysis validated conjugation of these glycans to human proteins, while quantitative proteomics monitored the glycoenzyme expression levels during macrophage differentiation. Significant interperson glycome variations were observed suggesting a considerable physiology-dependent or heritable heterogeneity of CD14+ monocytes. Only few N-glycome changes correlated with the monocyte-to-macrophage transition across donors including decreased core fucosylation and reduced expression of mannose-terminating (paucimannosidic-/oligomannosidic-type) N-glycans in macrophages, while lectin flow cytometry indicated that more dramatic cell surface glycan remodelling occurs during maturation. The less heterogeneous core 1-rich O-glycome showed a minor decrease in core 2-type O-glycosylation but otherwise remained unchanged with macrophage maturation. This high-resolution glycome map underpinning normal monocyte-to-macrophage transition, the most detailed to date, aids our understanding of the molecular makeup pertaining to two vital innate immune cell types and forms an important reference for future glycoimmunological studies.
Collapse
Affiliation(s)
- Hannes Hinneburg
- Department of Molecular Sciences, Macquarie University, NSW 2109 Sydney, Australia
- Biomolecular Discovery Research Centre, Macquarie University, NSW 2109 Sydney, Australia
| | - Jessica L Pedersen
- School of Life Sciences, Faculty of Science, University of Technology, NSW 2007 Sydney, Australia
| | - Nilesh J Bokil
- School of Life Sciences, Faculty of Science, University of Technology, NSW 2007 Sydney, Australia
| | - Alexander Pralow
- Max Planck Institute for Dynamics of Complex Technical Systems (Bioprocess Engineering), 39106 Magdeburg, Germany
| | | | - Rebeca Kawahara
- Department of Molecular Sciences, Macquarie University, NSW 2109 Sydney, Australia
- Biomolecular Discovery Research Centre, Macquarie University, NSW 2109 Sydney, Australia
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems (Bioprocess Engineering), 39106 Magdeburg, Germany
- GlyXera GmbH, 39120 Magdeburg, Germany
| | - Bernadette M Saunders
- School of Life Sciences, Faculty of Science, University of Technology, NSW 2007 Sydney, Australia
| | - Morten Thaysen-Andersen
- Department of Molecular Sciences, Macquarie University, NSW 2109 Sydney, Australia
- Biomolecular Discovery Research Centre, Macquarie University, NSW 2109 Sydney, Australia
| |
Collapse
|
18
|
Sukumaran A, Coish J, Yeung J, Muselius B, Gadjeva M, MacNeil A, Geddes-McAlister J. Decoding communication patterns of the innate immune system by quantitative proteomics. J Leukoc Biol 2019; 106:1221-1232. [PMID: 31556465 PMCID: PMC7309189 DOI: 10.1002/jlb.2ri0919-302r] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/12/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022] Open
Abstract
The innate immune system is a collective network of cell types involved in cell recruitment and activation using a robust and refined communication system. Engagement of receptor-mediated intracellular signaling initiates communication cascades by conveying information about the host cell status to surrounding cells for surveillance and protection. Comprehensive profiling of innate immune cells is challenging due to low cell numbers, high dynamic range of the cellular proteome, low abundance of secreted proteins, and the release of degradative enzymes (e.g., proteases). However, recent advances in mass spectrometry-based proteomics provides the capability to overcome these limitations through profiling the dynamics of cellular processes, signaling cascades, post-translational modifications, and interaction networks. Moreover, integration of technologies and molecular datasets provide a holistic view of a complex and intricate network of communications underscoring host defense and tissue homeostasis mechanisms. In this Review, we explore the diverse applications of mass spectrometry-based proteomics in innate immunity to define communication patterns of the innate immune cells during health and disease. We also provide a technical overview of mass spectrometry-based proteomic workflows, with a focus on bottom-up approaches, and we present the emerging role of proteomics in immune-based drug discovery while providing a perspective on new applications in the future.
Collapse
Affiliation(s)
- A. Sukumaran
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - J.M. Coish
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1
| | - J. Yeung
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - B. Muselius
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON, Canada, N1G 2W1
| | - M. Gadjeva
- Division of Infectious Diseases, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA 02115
| | - A.J. MacNeil
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1
| | - J. Geddes-McAlister
- Molecular and Cellular Biology Department, University of Guelph, Guelph, ON, Canada, N1G 2W1
| |
Collapse
|
19
|
Roos A, Preusse C, Hathazi D, Goebel HH, Stenzel W. Proteomic Profiling Unravels a Key Role of Specific Macrophage Subtypes in Sporadic Inclusion Body Myositis. Front Immunol 2019; 10:1040. [PMID: 31143183 PMCID: PMC6522546 DOI: 10.3389/fimmu.2019.01040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 04/23/2019] [Indexed: 12/14/2022] Open
Abstract
Unbiased proteomic profiling was performed toward the identification of biological parameters relevant in sIBM, thus giving hints about the pathophysiological processes and the existence of new reliable markers. For that purpose, skeletal muscle biopsies from 13 sIBM and 7 non-diseased control patients were analyzed with various methods, including liquid chromatography coupled to tandem mass spectrometry (four patients). Subsequent data analysis identified key molecules further studied in a larger cohort by qPCR, immunostaining, and immunofluorescence in situ. Proteomic signature of muscle biopsies derived from sIBM patients revealed the chaperone and cell surface marker CD74, the macrophage scavenger molecule CD163 and the transcription activator STAT1 to be among the highly and relevantly expressed proteins suggesting a significant contribution of immune cells among the myofibers expressing these markers. Moreover, in silico studies showed that 39% of upregulated proteins were involved in type I or mixed type I and type II interferon immunity. Indeed, further studies via immunohistochemistry clearly confirmed the prominent involvement of the key type I interferon signature-related molecules, ISG15 as well as IRF8 with MHC class II+ myofibers. Siglec1 colocalized with CD163+ macrophages and MHC class II molecules also co-localized with CD74 on macrophages. STAT1 co-localized with Siglec1+ macrophages in active myofibre myophagocytosis while STAT6 colocalized with endomysial macrophages. These combined results show involvement of CD74, CD163, and STAT1 as key molecules of macrophage activation being crucially involved in mixed and specific type I interferon, and interferon gamma associated-pathways in sIBM. On a more general note, these results also highlight the type of immune-interaction between macrophages and myofibers in the etiopathology of sIBM.
Collapse
Affiliation(s)
- Andreas Roos
- Department of Neuropediatrics, Developmental Neurology and Social Pediatrics, Centre for Neuromuscular Disorders in Children, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Corinna Preusse
- Department of Neuropathology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Denisa Hathazi
- Leibniz-Institut für Analytische Wissenschaften -ISAS- e.V., Dortmund, Germany
| | - Hans-Hilmar Goebel
- Department of Neuropathology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité -Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Leibniz Science Campus Chronic Inflammation, Berlin, Germany
| |
Collapse
|
20
|
Koo SJ, Garg NJ. Metabolic programming of macrophage functions and pathogens control. Redox Biol 2019; 24:101198. [PMID: 31048245 PMCID: PMC6488820 DOI: 10.1016/j.redox.2019.101198] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 04/09/2019] [Indexed: 12/15/2022] Open
Abstract
Macrophages (Mφ) are central players in mediating proinflammatory and immunomodulatory functions. Unchecked Mφ activities contribute to pathology across many diseases, including those caused by infectious pathogens and metabolic disorders. A fine balance of Mφ responses is crucial, which may be achieved by enforcing appropriate bioenergetics pathways. Metabolism serves as the provider of energy, substrates, and byproducts that support differential Mφ characteristics. The metabolic properties that control the polarization and response of Mφ remain to be fully uncovered for use in managing infectious diseases. Here, we review the various metabolic states in Mφ and how they influence the cell function.
Collapse
Affiliation(s)
- Sue-Jie Koo
- Department of Pathology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Nisha J Garg
- Department of Microbiology & Immunology, UTMB, Galveston, TX, USA; Institute for Human Infections and Immunity, UTMB, Galveston, TX, USA.
| |
Collapse
|
21
|
3D type I collagen environment leads up to a reassessment of the classification of human macrophage polarizations. Biomaterials 2019; 208:98-109. [PMID: 31005702 DOI: 10.1016/j.biomaterials.2019.04.018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/21/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Macrophages have multiple roles in development, tissue homeostasis and repair and present a high degree of phenotypic plasticity embodied in the concept of polarization. One goal of macrophage biology field is to characterize these polarizations at the molecular level. To achieve this task, it is necessary to integrate how physical environment signals are interpreted by macrophages under immune stimulation. In this work, we study how a 3D scaffold obtained from polymerized fibrillar rat type I collagen modulates the polarizations of human macrophages and reveal that some traditionally used markers should be reassessed. We demonstrate that integrin β2 is a regulator of STAT1 phosphorylation in response to IFNγ/LPS as well as responsible for the inhibition of ALOX15 expression in response to IL-4/IL-13 in 3D. Meanwhile, we also find that the CCL19/CCL20 ratio is reverted in 3D under IFNγ/LPS stimulation. 3D also induces the priming of the NLRP3 inflammasome resulting in an increased IL-1β and IL-6 secretion. These results give the molecular basis for assessing collagen induced immunomodulation of human macrophages in various physiological and pathological contexts such as cancer.
Collapse
|
22
|
Court M, Barnes JP, Millet A. Identifying exposition to low oxygen environment in human macrophages using secondary ion mass spectrometry and multivariate analysis. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2017; 31:1623-1632. [PMID: 28755479 DOI: 10.1002/rcm.7946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 06/07/2023]
Abstract
RATIONALE Macrophages are innate immune cells presenting a strong phenotypic plasticity and deeply involved in tissue homeostasis. Oxygen environmental tension is a physical parameter that could influence their polarizations. In this study we use time-of-flight secondary ion mass spectrometry (TOF-SIMS) to describe how various polarizations are modified by a low oxygen exposure. METHODS TOF-SIMS experiments were performed using an IONTOF ToF-SIMS 5-100 (ION-TOF GmbH, Munster, Germany). Analysis was performed using a pulsed 25 keV Bi3+ beam, sputtering was performed using a 250 eV Cs beam. Cells were fixed by paraformaldehyde before TOF-SIMS analysis. RESULTS Multivariate analysis of the TOF-SIMS spectra provided ion species associated with the exposure of macrophages to low oxygen concentration. We were able to obtain some species, specific of a particular polarization, advocating for the use of macrophages as reporter cells of oxygen tension in tissues. CONCLUSIONS Our study demonstrates that macrophage molecular signature to low oxygen environment is dependent on their polarization. TOF-SIMS shows the clear capability to produce species revealing this exposition. This result opens the way to the use of TOF-SIMS as a tool to explore hypoxia in human tissues.
Collapse
Affiliation(s)
- Magali Court
- Inserm U1205, Grenoble, France
- University of Grenoble-Alpes, Grenoble, France
| | - Jean-Paul Barnes
- University Grenoble Alpes, F-38000, Grenoble, France
- CEA, LETI, MINATEC Campus, F-38054, Grenoble, France
| | - Arnaud Millet
- Inserm U1205, Grenoble, France
- University of Grenoble-Alpes, Grenoble, France
- Team ATIP/Avenir Mechanobiology, Immunity and Cancer, Grenoble, France
| |
Collapse
|