1
|
Leek C, Cantu A, Sonti S, Gutierrez MC, Eldredge L, Sajti E, Xu HN, Lingappan K. Role of sex as a biological variable in neonatal alveolar macrophages. Redox Biol 2024; 75:103296. [PMID: 39098263 PMCID: PMC11345582 DOI: 10.1016/j.redox.2024.103296] [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: 05/29/2024] [Revised: 07/09/2024] [Accepted: 07/31/2024] [Indexed: 08/06/2024] Open
Abstract
The lung macrophages play a crucial role in health and disease. Sexual dimorphism significantly impacts the phenotype and function of tissue-resident macrophages. The primary mechanisms responsible for sexually dimorphic outcomes in bronchopulmonary dysplasia (BPD) remain unidentified. We tested the hypothesis that biological sex plays a crucial role in the transcriptional state of alveolar macrophages, using neonatal murine hyperoxia-induced lung injury as a relevant model for human BPD. The effects of neonatal hyperoxia exposure (95 % FiO2, PND1-5: saccular stage) on the lung myeloid cells acutely after injury and during normoxic recovery were measured. Alveolar macrophages (AM) from room air- and hyperoxia exposed from male and female neonatal murine lungs were subjected to bulk-RNA Sequencing. AMs are significantly depleted in the hyperoxia-exposed lung acutely after injury, with subsequent recovery in both sexes. The transcriptome of the alveolar macrophages is impacted by neonatal hyperoxia exposure and by sex as a biological variable. Pathways related to DNA damage and interferon-signaling were positively enriched in female AMs. Metabolic pathways related to glucose and carbohydrate metabolism were positively enriched in the male AMs, while oxidative phosphorylation was negatively enriched. These pathways were shared with monocytes and airway macrophages from intubated male and female human premature neonates.
Collapse
Affiliation(s)
- Connor Leek
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Abiud Cantu
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Shilpa Sonti
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Manuel Cantu Gutierrez
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA
| | - Laurie Eldredge
- Department of Pediatrics, Division of Pediatric Pulmonology, University of Washington School of Medicine, Seattle Children's Hospital, WA, USA
| | - Eniko Sajti
- Department of Pediatrics, Division of Neonatology, University of California San Diego, San Diego, CA, USA
| | - He N Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Krithika Lingappan
- Department of Pediatrics, Division of Neonatology, Children's Hospital of Philadelphia, University of Pennsylvania, PA, USA.
| |
Collapse
|
2
|
Kong L, Sun P, Pan X, Xiao C, Song B, Song Z. Glycerol monolaurate regulates apoptosis and inflammation by suppressing lipopolysaccharide-induced ROS production and NF-κB activation in avian macrophages. Poult Sci 2024; 103:103870. [PMID: 38851181 PMCID: PMC11208948 DOI: 10.1016/j.psj.2024.103870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 06/10/2024] Open
Abstract
Macrophages play a crucial role in both innate and adaptive immunity. However, their abnormal activation can lead to undesirable inflammatory reactions. This study aimed to investigate the effects of glycerol monolaurate (GML), a natural monoester known for its anti-inflammatory and immunoregulatory properties, on avian macrophages using the HD11 cell line. The results indicated that a concentration of 10 μg/mL of GML enhanced the phagocytic activity of HD11 cells (P < 0.05) without affecting cell viability (P > 0.05). GML decreased the expression of M1 macrophage polarization markers, such as CD86 and TNF-α genes (P < 0.05), while increasing the expression of M2 macrophage polarization markers, such as TGF-β1 and IL-10 genes (P < 0.05). GML suppressed ROS production, apoptosis, and the expression of proinflammatory genes (IL-1β and IL-6) induced by LPS (P < 0.05). GML also promoted the expression of TGF-β1 and IL-10 (P < 0.05), both in the presence and absence of LPS exposure. Moreover, GML suppressed the gene expression of TLR4 and NF-κB p65 induced by LPS (P < 0.05), as well as the phosphorylation of NF-κB p65 (P < 0.05). In conclusion, GML exhibited regulatory effects on the polarized state of avian macrophages and demonstrated significant anti-apoptotic and anti-inflammatory properties by suppressing intracellular ROS and the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Linglian Kong
- Office of Assessment, Jining Polytechnic, Jining, Shandong 272037, China; Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Peng Sun
- Laboratory of Chemistry of Natural Molecules, Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Xue Pan
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Chuanpi Xiao
- Laboratory of Chemistry of Natural Molecules, Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Bochen Song
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China
| | - Zhigang Song
- Key Laboratory of Efficient Utilization of Non-grain Feed Resources, College of Animal Science and Technology, Shandong Agricultural University, Taian, Shandong 271018, China.
| |
Collapse
|
3
|
Ge Z, Chen Y, Ma L, Hu F, Xie L. Macrophage polarization and its impact on idiopathic pulmonary fibrosis. Front Immunol 2024; 15:1444964. [PMID: 39131154 PMCID: PMC11310026 DOI: 10.3389/fimmu.2024.1444964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/12/2024] [Indexed: 08/13/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a lung disease that worsens over time, causing fibrosis in the lungs and ultimately resulting in respiratory failure and a high risk of death. Macrophages play a crucial role in the immune system, showing flexibility by transforming into either pro-inflammatory (M1) or anti-inflammatory (M2) macrophages when exposed to different stimuli, ultimately impacting the development of IPF. Recent research has indicated that the polarization of macrophages is crucial in the onset and progression of IPF. M1 macrophages secrete inflammatory cytokines and agents causing early lung damage and fibrosis, while M2 macrophages support tissue healing and fibrosis by releasing anti-inflammatory cytokines. Developing novel treatments for IPF relies on a thorough comprehension of the processes involved in macrophage polarization in IPF. The review outlines the regulation of macrophage polarization and its impact on the development of IPF, with the goal of investigating the possible therapeutic benefits of macrophage polarization in the advancement of IPF.
Collapse
Affiliation(s)
- Zhouling Ge
- Department of Respiratory Medicine, The Third Affiliated Hospital of Shanghai University (Wenzhou People’s Hospital), Wenzhou, China
| | - Yong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Leikai Ma
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangjun Hu
- Department of Respiratory Medicine, The Third Affiliated Hospital of Shanghai University (Wenzhou People’s Hospital), Wenzhou, China
| | - Lubin Xie
- Department of Respiratory Medicine, The Third Affiliated Hospital of Shanghai University (Wenzhou People’s Hospital), Wenzhou, China
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
4
|
Zhao C, Yang Z, Li Y, Wen Z. Macrophages in tissue repair and regeneration: insights from zebrafish. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:12. [PMID: 38861103 PMCID: PMC11166613 DOI: 10.1186/s13619-024-00195-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/31/2024] [Indexed: 06/12/2024]
Abstract
Macrophages play crucial and versatile roles in regulating tissue repair and regeneration upon injury. However, due to their complex compositional heterogeneity and functional plasticity, deciphering the nature of different macrophage subpopulations and unraveling their dynamics and precise roles during the repair process have been challenging. With its distinct advantages, zebrafish (Danio rerio) has emerged as an invaluable model for studying macrophage development and functions, especially in tissue repair and regeneration, providing valuable insights into our understanding of macrophage biology in health and diseases. In this review, we present the current knowledge and challenges associated with the role of macrophages in tissue repair and regeneration, highlighting the significant contributions made by zebrafish studies. We discuss the unique advantages of the zebrafish model, including its genetic tools, imaging techniques, and regenerative capacities, which have greatly facilitated the investigation of macrophages in these processes. Additionally, we outline the potential of zebrafish research in addressing the remaining challenges and advancing our understanding of the intricate interplay between macrophages and tissue repair and regeneration.
Collapse
Affiliation(s)
- Changlong Zhao
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhiyong Yang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Yunbo Li
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
- Division of Life Science, the Hong Kong University of Science and Technology, Clear Water Bay, Hong Kong, China
| | - Zilong Wen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
5
|
Holt M, Lin J, Cicka M, Wong A, Epelman S, Lavine KJ. Dissecting and Visualizing the Functional Diversity of Cardiac Macrophages. Circ Res 2024; 134:1791-1807. [PMID: 38843293 DOI: 10.1161/circresaha.124.323817] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024]
Abstract
Cardiac macrophages represent a functionally diverse population of cells involved in cardiac homeostasis, repair, and remodeling. With recent advancements in single-cell technologies, it is possible to elucidate specific macrophage subsets based on transcriptional signatures and cell surface protein expression to gain a deep understanding of macrophage diversity in the heart. The use of fate-mapping technologies and parabiosis studies have provided insight into the ontogeny and dynamics of macrophages identifying subsets derived from embryonic and adult definitive hematopoietic progenitors that include tissue-resident and bone marrow monocyte-derived macrophages, respectively. Within the heart, these subsets have distinct tissue niches and functional roles in the setting of homeostasis and disease, with cardiac resident macrophages representing a protective cell population while bone marrow monocyte-derived cardiac macrophages have a context-dependent effect, triggering both proinflammatory tissue injury, but also promoting reparative functions. With the increased understanding of the clinical relevance of cardiac macrophage subsets, there has been an increasing need to detect and measure cardiac macrophage compositions in living animals and patients. New molecular tracers compatible with positron emission tomography/computerized tomography and positron emission tomography/ magnetic resonance imaging have enabled investigators to noninvasively and serially visualize cardiac macrophage subsets within the heart to define associations with disease and measure treatment responses. Today, advancements within this thriving field are poised to fuel an era of clinical translation.
Collapse
Affiliation(s)
- Megan Holt
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| | - Julia Lin
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
| | - Markus Cicka
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| | - Anthony Wong
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada (J.L., A.W., S.E.)
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada (S.E.)
- Department of Immunology, University of Toronto, ON, Canada (J.L., A.W., S.E.)
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada (S.E.)
| | - Kory J Lavine
- Division of Cardiology, Department of Medicine, Center for Cardiovascular Research, Washington University School of Medicine (M.H., M.C., K.J.L.)
| |
Collapse
|
6
|
Lim PN, Cervantes MM, Pham LK, Doherty S, Tufts A, Dubey D, Mai D, Aderem A, Diercks AH, Rothchild AC. Absence of c-Maf and IL-10 enables Type I IFN enhancement of innate responses to low-dose LPS in alveolar macrophages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.594428. [PMID: 38826239 PMCID: PMC11142172 DOI: 10.1101/2024.05.22.594428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Alveolar macrophages (AMs) are lower-airway resident myeloid cells and are among the first to respond to inhaled pathogens. Here, we interrogate AM innate sensing to Pathogen Associated Molecular Patterns (PAMPs) and determine AMs have decreased responses to low-dose LPS compared to other macrophages, as measured by TNF, IL-6, Ifnb, and Ifit3. We find the reduced response to low-dose LPS correlates with minimal TLR4 and CD14 surface expression, despite sufficient internal expression of TLR4. Additionally, we find that AMs do not produce IL-10 in response to a variety of PAMPs due to low expression of transcription factor c-Maf and that lack of IL-10 production contributes to an enhancement of pro-inflammatory responses by Type I IFN. Our findings demonstrate that AMs have cell-intrinsic dampened responses to LPS, which is enhanced by type I IFN exposure. These data implicate conditions where AMs may have reduced or enhanced sentinel responses to bacterial infections.
Collapse
Affiliation(s)
- Pamelia N. Lim
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Maritza M. Cervantes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Linh K. Pham
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Graduate Program in Animal Biotechnology & Biomedical Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Sydney Doherty
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Ankita Tufts
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| | - Divya Dubey
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Dat Mai
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98019
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98019
| | - Alan H. Diercks
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA 98019
| | - Alissa C. Rothchild
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|
7
|
Yi W, Lv D, Sun Y, Mu J, Lu X. Role of APOE in glaucoma. Biochem Biophys Res Commun 2024; 694:149414. [PMID: 38145596 DOI: 10.1016/j.bbrc.2023.149414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 12/27/2023]
Abstract
Glaucoma is a chronic blinding eye disease caused by the progressive loss of retinal ganglion cells (RGCs). Currently, no clinically approved treatment can directly improve the survival rate of RGCs. The Apolipoprotein E (APOE) gene is closely related to the genetic risk of numerous neurodegenerative diseases and has become a hot topic in the field of neurodegenerative disease research in recent years. The optic nerve and retina are extensions of the brain's nervous system. The pathogenesis of retinal degenerative diseases is closely related to the degenerative diseases of the nerves in the brain. APOE consists of three alleles, ε4, ε3, and ε2, in a single locus. They have varying degrees of risk for glaucoma. APOE4 and the APOE gene deletion (APOE-/-) can reduce RGC loss. By contrast, APOE3 and the overall presence of APOE genes (APOE+/+) result in significant loss of RGC bodies and axons, increasing the risk of glaucoma RGCs death. Currently, there is no clear literature indicating that APOE2 is beneficial or harmful to glaucoma. This study summarises the mechanism of different APOE genes in glaucoma and speculates that APOE targeted intervention may be a promising method for protecting against RGCs loss in glaucoma.
Collapse
Affiliation(s)
- Wenhua Yi
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - De Lv
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, China.
| | - Yue Sun
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - Jingyu Mu
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China.
| | - Xuejing Lu
- Eye School of Chengdu University of TCM, Chengdu City, Sichuan province, China; Ineye Hospital of Chengdu University of TCM, Chengdu City, Sichuan province, China; Key Laboratory of Sichuan Province Ophthalmopathy Prevention & Cure and Visual Function Protection with TCM Laboratory, Chengdu City, Sichuan province, China; Retinal Image Technology and Chronic Vascular Disease Prevention&Control and Collaborative Innovation Center, Chengdu City, Sichuan province, China.
| |
Collapse
|
8
|
Jannini-Sá YAP, Creyns B, Hogaboam CM, Parks WC, Hohmann MS. Macrophages in Lung Repair and Fibrosis. Results Probl Cell Differ 2024; 74:257-290. [PMID: 39406909 DOI: 10.1007/978-3-031-65944-7_10] [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: 11/01/2024]
Abstract
Macrophages are key regulators of tissue repair and fibrosis. Following injury, macrophages undergo marked phenotypic and functional changes to play crucial roles throughout the phases of tissue repair. Idiopathic Pulmonary Fibrosis, which is the most common fibrosing lung disease, has been described as an aberrant reparative response to repetitive alveolar epithelial injury in a genetically susceptible aging individual. The marked destruction of the lung architecture results from the excessive secretion of extracellular matrix by activated fibroblasts and myofibroblasts. Accumulating evidence suggests that macrophages have a pivotal regulatory role in pulmonary fibrosis. The origins and characteristics of macrophages in the lung and their role in regulating lung homeostasis, repair, and fibrosis are reviewed herein. We discuss recent studies that have employed single-cell RNA-sequencing to improve the identification and characterization of macrophage populations in the context of homeostatic and fibrotic conditions. We also discuss the current understanding of the macrophage-mediated mechanisms underlying the initiation and progression of pulmonary fibrosis, with a focus on the phenotypic and functional changes that aging macrophages acquire and how these changes ultimately contribute to age-related chronic lung diseases.
Collapse
Affiliation(s)
- Yago A P Jannini-Sá
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Brecht Creyns
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cory M Hogaboam
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - William C Parks
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Miriam S Hohmann
- Women's Guild Lung Institute, Division of Pulmonary & Critical Care Medicine, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Welfley H, Kylat R, Zaghloul N, Halonen M, Martinez FD, Ahmed M, Cusanovich DA. Single-Cell Profiling of Premature Neonate Airways Reveals a Continuum of Myeloid Differentiation. Am J Respir Cell Mol Biol 2023; 69:689-697. [PMID: 37643399 PMCID: PMC10704120 DOI: 10.1165/rcmb.2022-0293oc] [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: 07/28/2023] [Accepted: 08/28/2023] [Indexed: 08/31/2023] Open
Abstract
Single-cell genomic technologies hold great potential to advance our understanding of lung development and disease. A major limitation lies in accessing intact cells from primary lung tissues for profiling human airway health. Sampling methods such as endotracheal aspiration that are compatible with clinical interventions could enable longitudinal studies, the enrollment of large cohorts, and the development of novel diagnostics. To explore single-cell RNA sequencing profiling of the cell types present at birth in the airway lumen of extremely premature neonates (<28 wk gestation), we isolated cells from endotracheal aspirates collected from intubated neonates within the first hour after birth. We generated data on 10 subjects, providing a rich view of airway luminal biology at a critical developmental period. Our results show that cells present in the airways of premature neonates primarily represent a continuum of myeloid differentiation, including fetal monocytes (25% of total), intermediate myeloid populations (48%), and macrophages (2.6%). Applying trajectory analysis to the myeloid populations, we identified two trajectories consistent with the developmental stages of interstitial and alveolar macrophages, as well as a third trajectory presenting an alternative pathway bridging the distinct macrophage precursors. The three trajectories share many dynamic genes (N = 5,451), but also have distinct transcriptional changes (259 alveolar-specific, 666 interstitial-specific, and 285 bridging-specific). Overall, our results define cells isolated within the so-called "golden hour of birth" in extremely premature neonate airways, representing complex lung biology, and can be used in studies of human development and disease.
Collapse
Affiliation(s)
| | - Ranjit Kylat
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona College of Medicine, Tucson, Arizona
| | - Nahla Zaghloul
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona College of Medicine, Tucson, Arizona
| | | | | | - Mohamed Ahmed
- Department of Pediatrics, Steele Children’s Research Center, University of Arizona College of Medicine, Tucson, Arizona
| | - Darren A. Cusanovich
- Asthma and Airway Disease Research Center and
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona; and
| |
Collapse
|
10
|
Chen J, Zhao X, Huang C, Lin J. Novel insights into molecular signatures and pathogenic cell populations shared by systemic lupus erythematosus and vascular dementia. Funct Integr Genomics 2023; 23:337. [PMID: 37971684 DOI: 10.1007/s10142-023-01270-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/19/2023]
Abstract
Although vascular dementia (VD) and systemic lupus erythematosus (SLE) may share immune-mediated pathophysiologic processes, the underlying mechanisms are unclear. This study investigated shared gene signatures in SLE versus VD, as well as their potential molecular mechanisms. Bulk RNA sequencing (RNAseq) and single-cell or single-nucleus RNAseq (sc/snRNAseq) datasets from SLE blood samples and VD brain samples were obtained from Gene Expression Omnibus. The identification of genes associated with both SLE and VD was performed using the weighted gene co-expression network analysis (WGCNA) and machine learning algorithms. For the sc/snRNAseq data, an unbiased clustering pipeline based on Seurat and CellChat was used to determine the cellular landscape profile and examine intracellular communication, respectively. The results were subsequently validated using a mice model of SLE with cognitive dysfunction (female MRL/lpr mice). WGCNA and machine learning identified C1QA, LY96, CD163, and MS4A4A as key genes for SLE and VD. sc/snRNAseq analyses revealed that CD163 and MS4A4A were upregulated in mononuclear phagocytes (MPs) from SLE and VD samples and were associated with monocyte-macrophage differentiation. Intriguingly, LGALS9-associated molecular pathway, as the only signaling pathway common between SLE and VD via CellChat analysis, exhibited significant upregulation in cortical microglia of MRL/lpr mice. Our analyses identified C1QA, LY96, CD163, and MS4A4A as potential biomarkers for SLE and VD. Moreover, the upregulation of CD163/MS4A4A and activation of LGALS9 signaling in MPs may contribute to the pathogenesis of VD with SLE. These findings offer novel insight into the mechanisms underlying VD in SLE patients.
Collapse
Affiliation(s)
- Jing Chen
- Department of Neurology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510630, China
- Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xiao'feng Zhao
- Department of Rheumatology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Cheng Huang
- Department of Neurology, Clinical Neuroscience Institute, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jia'xing Lin
- Department of Neurology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510630, China.
| |
Collapse
|
11
|
Thomas SK, Wattenberg MM, Choi-Bose S, Uhlik M, Harrison B, Coho H, Cassella CR, Stone ML, Patel D, Markowitz K, Delman D, Chisamore M, Drees J, Bose N, Beatty GL. Kupffer cells prevent pancreatic ductal adenocarcinoma metastasis to the liver in mice. Nat Commun 2023; 14:6330. [PMID: 37816712 PMCID: PMC10564762 DOI: 10.1038/s41467-023-41771-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 09/12/2023] [Indexed: 10/12/2023] Open
Abstract
Although macrophages contribute to cancer cell dissemination, immune evasion, and metastatic outgrowth, they have also been reported to coordinate tumor-specific immune responses. We therefore hypothesized that macrophage polarization could be modulated therapeutically to prevent metastasis. Here, we show that macrophages respond to β-glucan (odetiglucan) treatment by inhibiting liver metastasis. β-glucan activated liver-resident macrophages (Kupffer cells), suppressed cancer cell proliferation, and invoked productive T cell-mediated responses against liver metastasis in pancreatic cancer mouse models. Although excluded from metastatic lesions, Kupffer cells were critical for the anti-metastatic activity of β-glucan, which also required T cells. Furthermore, β-glucan drove T cell activation and macrophage re-polarization in liver metastases in mice and humans and sensitized metastatic lesions to anti-PD1 therapy. These findings demonstrate the significance of macrophage function in metastasis and identify Kupffer cells as a potential therapeutic target against pancreatic cancer metastasis to the liver.
Collapse
Affiliation(s)
- Stacy K Thomas
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Max M Wattenberg
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shaanti Choi-Bose
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark Uhlik
- HiberCell Inc, Roseville, MN, USA
- OncXerna, Waltham, MA, USA
| | | | - Heather Coho
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher R Cassella
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Meredith L Stone
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dhruv Patel
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelly Markowitz
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Devora Delman
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Gregory L Beatty
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Al-Mudares F, Cantu Gutierrez M, Cantu A, Jiang W, Wang L, Dong X, Moorthy B, Sajti E, Lingappan K. Loss of growth differentiation factor 15 exacerbates lung injury in neonatal mice. Am J Physiol Lung Cell Mol Physiol 2023; 325:L314-L326. [PMID: 37368978 PMCID: PMC10625832 DOI: 10.1152/ajplung.00086.2023] [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: 03/21/2023] [Revised: 05/11/2023] [Accepted: 06/21/2023] [Indexed: 06/29/2023] Open
Abstract
Growth differentiation factor 15 (GDF15) is a divergent member of the transforming growth factor-β (TGF-β) superfamily, and its expression increases under various stress conditions, including inflammation, hyperoxia, and senescence. GDF15 expression is increased in neonatal murine bronchopulmonary dysplasia (BPD) models, and GDF15 loss exacerbates oxidative stress and decreases cellular viability in vitro. Our overall hypothesis is that the loss of GDF15 will exacerbate hyperoxic lung injury in the neonatal lung in vivo. We exposed neonatal Gdf15-/- mice and wild-type (WT) controls on a similar background to room air or hyperoxia (95% [Formula: see text]) for 5 days after birth. The mice were euthanized on postnatal day 21 (PND 21). Gdf15-/- mice had higher mortality and lower body weight than WT mice after exposure to hyperoxia. Hyperoxia exposure adversely impacted alveolarization and lung vascular development, with a greater impact in Gdf15-/- mice. Interestingly, Gdf15-/- mice showed lower macrophage count in the lungs compared with WT mice both under room air and after exposure to hyperoxia. Analysis of the lung transcriptome revealed marked divergence in gene expression and enriched biological pathways in WT and Gdf15-/- mice and differed markedly by biological sex. Notably, pathways related to macrophage activation and myeloid cell homeostasis were negatively enriched in Gdf15-/- mice. Loss of Gdf15 exacerbates mortality, lung injury, and the phenotype of the arrest of alveolarization in the developing lung with loss of female-sex advantage in Gdf15-/- mice.NEW & NOTEWORTHY We show for the first time that loss of Gdf15 exacerbates mortality, lung injury, and the phenotype of the arrest of alveolarization in the developing lung with loss of female-sex advantage in Gdf15-/- mice. We also highlight the distinct pulmonary transcriptomic response in the Gdf15-/- lung including pathways related to macrophage recruitment and activation.
Collapse
Affiliation(s)
- Faeq Al-Mudares
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Manuel Cantu Gutierrez
- Divsion of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Abiud Cantu
- Divsion of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Weiwu Jiang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Lihua Wang
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Xiaoyu Dong
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Bhagavatula Moorthy
- Section of Neonatology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas, United States
| | - Eniko Sajti
- Division of Neonatology, Department of Pediatrics, University of California, San Diego, California, United States
| | - Krithika Lingappan
- Divsion of Neonatology, Department of Pediatrics, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| |
Collapse
|
13
|
Zhang S, Wen X, Zhou L, Li H, Li W, Hoffman AR, Hu JF, Cui J. Chromatin-RNA in situ Reverse Transcription Sequencing (CRIST-seq) Approach to Profile the Non-coding RNA Interaction Network. Bio Protoc 2023; 13:e4718. [PMID: 37497457 PMCID: PMC10366681 DOI: 10.21769/bioprotoc.4718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/02/2023] [Accepted: 04/28/2023] [Indexed: 07/28/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are defined as RNAs that do not encode proteins, but many ncRNAs do have the ability to regulate gene expression. These ncRNAs play a critical role in the epigenetic regulation of various physiological and pathological processes through diverse biochemical mechanisms. However, the existing screening methods to identify regulatory ncRNAs only focus on whole-cell expression levels and do not capture every ncRNA that targets certain genes. We describe a new method, chromatin-RNA in situ reverse transcription sequencing (CRIST-seq), that can identify all the ncRNAs that are associated with the regulation of any given gene. In this article, we targeted the ncRNAs that are associated with pluripotent gene Sox2, allowing us to catalog the ncRNA regulation network of pluripotency maintenance. This methodology is universally applicable for the study of epigenetic regulation of any genes by making simple changes on the CRISPR-dCas9 gRNAs. Key features This method provides a new technique for screening ncRNAs and establishing chromatin interaction networks. The target gene for this method can be any gene of interest and any site in the entire genome. This method can be further extended to detect RNAs, DNAs, and proteins that interact with target genes. Graphical overview.
Collapse
Affiliation(s)
- Shilin Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
- Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Xue Wen
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Lei Zhou
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Hui Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Wei Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| | - Andrew R. Hoffman
- Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Ji-Fan Hu
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
- Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Jiuwei Cui
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, Cancer Center, First Hospital of Jilin University, Changchun, Jilin 130061, China
| |
Collapse
|
14
|
Fixsen BR, Han CZ, Zhou Y, Spann NJ, Saisan P, Shen Z, Balak C, Sakai M, Cobo I, Holtman IR, Warden AS, Ramirez G, Collier JG, Pasillas MP, Yu M, Hu R, Li B, Belhocine S, Gosselin D, Coufal NG, Ren B, Glass CK. SALL1 enforces microglia-specific DNA binding and function of SMADs to establish microglia identity. Nat Immunol 2023; 24:1188-1199. [PMID: 37322178 PMCID: PMC10307637 DOI: 10.1038/s41590-023-01528-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 05/04/2023] [Indexed: 06/17/2023]
Abstract
Spalt-like transcription factor 1 (SALL1) is a critical regulator of organogenesis and microglia identity. Here we demonstrate that disruption of a conserved microglia-specific super-enhancer interacting with the Sall1 promoter results in complete and specific loss of Sall1 expression in microglia. By determining the genomic binding sites of SALL1 and leveraging Sall1 enhancer knockout mice, we provide evidence for functional interactions between SALL1 and SMAD4 required for microglia-specific gene expression. SMAD4 binds directly to the Sall1 super-enhancer and is required for Sall1 expression, consistent with an evolutionarily conserved requirement of the TGFβ and SMAD homologs Dpp and Mad for cell-specific expression of Spalt in the Drosophila wing. Unexpectedly, SALL1 in turn promotes binding and function of SMAD4 at microglia-specific enhancers while simultaneously suppressing binding of SMAD4 to enhancers of genes that become inappropriately activated in enhancer knockout microglia, thereby enforcing microglia-specific functions of the TGFβ-SMAD signaling axis.
Collapse
Affiliation(s)
- Bethany R Fixsen
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Claudia Z Han
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Yi Zhou
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Nathanael J Spann
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Payam Saisan
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Zeyang Shen
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Christopher Balak
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Mashito Sakai
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Isidoro Cobo
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Inge R Holtman
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen and University Medical Center Groningen, Groningen, the Netherlands
| | - Anna S Warden
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | | | - Jana G Collier
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Martina P Pasillas
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Miao Yu
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Rong Hu
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Bin Li
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Sarah Belhocine
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Quebec, Quebec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Quebec, Quebec, Canada
| | - David Gosselin
- Axe Neuroscience, Centre de Recherche du CHU de Québec, Université Laval, Quebec, Quebec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Quebec, Quebec, Canada
| | - Nicole G Coufal
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, UC San Diego, La Jolla, CA, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA
- Ludwig Institute for Cancer Research, La Jolla, CA, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
- Department of Medicine, School of Medicine, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
15
|
Cao C, Memet O, Liu F, Hu H, Zhang L, Jin H, Cao Y, Zhou J, Shen J. Transcriptional Characterization of Bronchoalveolar Lavage Fluid Reveals Immune Microenvironment Alterations in Chemically Induced Acute Lung Injury. J Inflamm Res 2023; 16:2129-2147. [PMID: 37220504 PMCID: PMC10200123 DOI: 10.2147/jir.s407580] [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: 02/22/2023] [Accepted: 05/10/2023] [Indexed: 05/25/2023] Open
Abstract
Purpose Chemically induced acute lung injury (CALI) has become a serious health concern in our industrialized world, and abnormal functional alterations of immune cells crucially contribute to severe clinical symptoms. However, the cell heterogeneity and functional phenotypes of respiratory immune characteristics related to CALI remain unclear. Methods We performed scRNA sequencing on bronchoalveolar lavage fluid (BALF) samples obtained from phosgene-induced CALI rat models and healthy controls. Transcriptional data and TotalSeq technology were used to confirm cell surface markers identifying immune cells in BALF. The landscape of immune cells could elucidate the metabolic remodeling mechanism involved in the progression of acute respiratory distress syndrome and cytokine storms. We used pseudotime inference to build macrophage trajectories and the corresponding model gene expression changes, and identified and characterized alveolar cells and immune subsets that may contribute to CALI pathophysiology based on gene expression profiles at single-cell resolution. Results The immune environment of cells, including dendritic cells and specific macrophage subclusters, exhibited increased function during the early stage of pulmonary tissue damage. Nine different subpopulations were identified that perform multiple functional roles, including immune responses, pulmonary tissue repair, cellular metabolic cycle, and cholesterol metabolism. Additionally, we found that individual macrophage subpopulations dominate the cell-cell communication landscape. Moreover, pseudo-time trajectory analysis suggested that proliferating macrophage clusters exerted multiple functional roles. Conclusion Our findings demonstrate that the bronchoalveolar immune microenvironment is a fundamental aspect of the immune response dynamics involved in the pathogenesis and recovery of CALI.
Collapse
Affiliation(s)
- Chao Cao
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, People’s Republic of China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People’s Republic of China
- Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Emergency Department, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Obulkasim Memet
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, People’s Republic of China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People’s Republic of China
- Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Fuli Liu
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, People’s Republic of China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People’s Republic of China
| | - Hanbing Hu
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, People’s Republic of China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People’s Republic of China
- Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| | - Lin Zhang
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, People’s Republic of China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People’s Republic of China
| | - Heng Jin
- Emergency Department, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Yiqun Cao
- Emergency Department, Tianjin Medical University General Hospital, Tianjin, People’s Republic of China
| | - Jian Zhou
- Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital of Fudan University, Shanghai, People’s Republic of China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Jie Shen
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, People’s Republic of China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, People’s Republic of China
- Shanghai Medical College, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
16
|
Rodriguez-Rodriguez L, Gillet L, Machiels B. Shaping of the alveolar landscape by respiratory infections and long-term consequences for lung immunity. Front Immunol 2023; 14:1149015. [PMID: 37081878 PMCID: PMC10112541 DOI: 10.3389/fimmu.2023.1149015] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/15/2023] [Indexed: 04/07/2023] Open
Abstract
Respiratory infections and especially viral infections, along with other extrinsic environmental factors, have been shown to profoundly affect macrophage populations in the lung. In particular, alveolar macrophages (AMs) are important sentinels during respiratory infections and their disappearance opens a niche for recruited monocytes (MOs) to differentiate into resident macrophages. Although this topic is still the focus of intense debate, the phenotype and function of AMs that recolonize the niche after an inflammatory insult, such as an infection, appear to be dictated in part by their origin, but also by local and/or systemic changes that may be imprinted at the epigenetic level. Phenotypic alterations following respiratory infections have the potential to shape lung immunity for the long-term, leading to beneficial responses such as protection against allergic airway inflammation or against other infections, but also to detrimental responses when associated with the development of immunopathologies. This review reports the persistence of virus-induced functional alterations in lung macrophages, and discusses the importance of this imprinting in explaining inter-individual and lifetime immune variation.
Collapse
|
17
|
Single-cell sequencing reveals that endothelial cells, EndMT cells and mural cells contribute to the pathogenesis of cavernous malformations. Exp Mol Med 2023; 55:628-642. [PMID: 36914857 PMCID: PMC10073145 DOI: 10.1038/s12276-023-00962-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/13/2022] [Accepted: 01/01/2023] [Indexed: 03/14/2023] Open
Abstract
Cavernous malformations (CMs) invading the central nervous system occur in ~0.16-0.4% of the general population, often resulting in hemorrhages and focal neurological deficits. Further understanding of disease mechanisms and therapeutic strategies requires a deeper knowledge of CMs in humans. Herein, we performed single-cell RNA sequencing (scRNA-seq) analysis on unselected viable cells from twelve human CM samples and three control samples. A total of 112,670 high-quality cells were clustered into 11 major cell types, which shared a number of common features in CMs harboring different genetic mutations. A new EC subpopulation marked with PLVAP was uniquely identified in lesions. The cellular ligand‒receptor network revealed that the PLVAP-positive EC subcluster was the strongest contributor to the ANGPT and VEGF signaling pathways in all cell types. The PI3K/AKT/mTOR pathway was strongly activated in the PLVAP-positive subcluster even in non-PIK3CA mutation carriers. Moreover, endothelial-to-mesenchymal transition (EndMT) cells were identified for the first time in CMs at the single-cell level, which was accompanied by strong immune activation. The transcription factor SPI1 was predicted to be a novel key driver of EndMT, which was confirmed by in vitro and in vivo studies. A specific fibroblast-like phenotype was more prevalent in lesion smooth muscle cells, hinting at the role of vessel reconstructions and repairs in CMs, and we also confirmed that TWIST1 could induce SMC phenotypic switching in vitro and in vivo. Our results provide novel insights into the pathomechanism decryption and further precise therapy of CMs.
Collapse
|
18
|
Manouchehri N, Salinas VH, Hussain RZ, Stüve O. Distinctive transcriptomic and epigenomic signatures of bone marrow-derived myeloid cells and microglia in CNS autoimmunity. Proc Natl Acad Sci U S A 2023; 120:e2212696120. [PMID: 36730207 PMCID: PMC9963604 DOI: 10.1073/pnas.2212696120] [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: 07/23/2022] [Accepted: 12/22/2022] [Indexed: 02/03/2023] Open
Abstract
In the context of autoimmunity, myeloid cells of the central nervous system (CNS) constitute an ontogenically heterogeneous population that includes yolk sac-derived microglia and infiltrating bone marrow-derived cells (BMC). We previously identified a myeloid cell subset in the brain and spinal cord that expresses the surface markers CD88 and CD317 and is associated with the onset and persistence of clinical disease in the murine model of the human CNS autoimmune disorder, experimental autoimmune encephalomyelitis (EAE). We employed an experimental platform utilizing single-cell transcriptomic and epigenomic profiling of bone marrow-chimeric mice to categorically distinguish BMC from microglia during CNS autoimmunity. Analysis of gene expression and chromosomal accessibility identified CD88+CD317+ myeloid cells in the CNS of EAE mice as originating from BMC and microglia. Interestingly, each cell lineage exhibited overlapping and unique gene expression patterns and transcription factor motifs that allowed their segregation. Our observations will facilitate determining pathogenic contributions of BMC and microglia in CNS autoimmune disease. Ultimately, this agnostic characterization of myeloid cells will be required for devising disease stage-specific and tissue-specific interventions for CNS inflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Navid Manouchehri
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Victor H. Salinas
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX75390
- Neurology Section, Veterans Affairs North Texas Health Care System, Dallas, TX75216
| | - Rehana Z. Hussain
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Olaf Stüve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX75390
- Neurology Section, Veterans Affairs North Texas Health Care System, Dallas, TX75216
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
19
|
Reyfman PA, Malsin ES, Khuder B, Joshi N, Gadhvi G, Flozak AS, Carns MA, Aren K, Goldberg IA, Kim S, Alexander M, Sporn PHS, Misharin AV, Budinger GS, Lam AP, Hinchcliff M, Gottardi CJ, Winter DR. A Novel MIP-1-Expressing Macrophage Subtype in BAL Fluid from Healthy Volunteers. Am J Respir Cell Mol Biol 2023; 68:176-185. [PMID: 36174229 PMCID: PMC9986555 DOI: 10.1165/rcmb.2021-0123oc] [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: 03/09/2021] [Accepted: 09/29/2022] [Indexed: 02/03/2023] Open
Abstract
Tissue availability remains an important limitation of single-cell genomic technologies for investigating cellular heterogeneity in human health and disease. BAL represents a minimally invasive approach to assessing an individual's lung cellular environment for diagnosis and research. However, the lack of high-quality, healthy lung reference data is a major obstacle to using single-cell approaches to study a plethora of lung diseases. Here, we performed single-cell RNA sequencing on over 40,000 cells isolated from the BAL of four healthy volunteers. Of the six cell types or lineages we identified, macrophages were consistently the most numerous across individuals. Our analysis confirmed the expression of marker genes defining cell types despite background signals because of the ambient RNA found in many single-cell studies. We assessed the variability of gene expression across macrophages and defined a distinct subpopulation of cells expressing a set of genes associated with Macrophage Inflammatory Protein 1 (MIP-1). RNA in situ hybridization and reanalysis of published lung single-cell data validated the presence of this macrophage subpopulation. Thus, our study characterizes lung macrophage heterogeneity in healthy individuals and provides a valuable resource for future studies to understand the lung environment in health and disease.
Collapse
Affiliation(s)
- Paul A. Reyfman
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Elizabeth S. Malsin
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Basil Khuder
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Nikita Joshi
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Gaurav Gadhvi
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Annette S. Flozak
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Mary A. Carns
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Kathleen Aren
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Isaac A. Goldberg
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Seokjo Kim
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Michael Alexander
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Peter H. S. Sporn
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Alexander V. Misharin
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - G.R. Scott Budinger
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Anna P. Lam
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Monique Hinchcliff
- Department of Internal Medicine, Section of Rheumatology, Allergy, and Immunology, Yale School of Medicine, New Haven, Connecticut
| | - Cara J. Gottardi
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| | - Deborah R. Winter
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois and
| |
Collapse
|
20
|
Integrated proteomic and transcriptomic landscape of macrophages in mouse tissues. Nat Commun 2022; 13:7389. [PMID: 36450731 PMCID: PMC9712610 DOI: 10.1038/s41467-022-35095-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Macrophages are involved in tissue homeostasis and are critical for innate immune responses, yet distinct macrophage populations in different tissues exhibit diverse gene expression patterns and biological processes. While tissue-specific macrophage epigenomic and transcriptomic profiles have been reported, proteomes of different macrophage populations remain poorly characterized. Here we use mass spectrometry and bulk RNA sequencing to assess the proteomic and transcriptomic patterns, respectively, of 10 primary macrophage populations from seven mouse tissues, bone marrow-derived macrophages and the cell line RAW264.7. The results show distinct proteomic landscape and protein copy numbers between tissue-resident and recruited macrophages. Construction of a hierarchical regulatory network finds cell-type-specific transcription factors of macrophages serving as hubs for denoting tissue and functional identity of individual macrophage subsets. Finally, Il18 is validated to be essential in distinguishing molecular signatures and cellular function features between tissue-resident and recruited macrophages in the lung and liver. In summary, these deposited datasets and our open proteome server ( http://macrophage.mouseprotein.cn ) integrating all information will provide a valuable resource for future functional and mechanistic studies of mouse macrophages.
Collapse
|
21
|
Gu Y, Lawrence T, Mohamed R, Liang Y, Yahaya BH. The emerging roles of interstitial macrophages in pulmonary fibrosis: A perspective from scRNA-seq analyses. Front Immunol 2022; 13:923235. [PMID: 36211428 PMCID: PMC9536737 DOI: 10.3389/fimmu.2022.923235] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
Pulmonary fibrosis is an irreversible and progressive disease affecting the lungs, and the etiology remains poorly understood. This disease can be lethal and currently has no specific clinical therapeutic regimen. Macrophages, the most common type of immune cell in the lungs, have been reported to play a key role in the pathogenesis of fibrotic disease. The lung macrophage population is mostly composed of alveolar macrophages and interstitial macrophages, both of which have not been thoroughly studied in the pathogenesis of lung fibrosis. Interstitial macrophages have recently been recognised for their participation in lung fibrosis due to new technology arising from a combination of bioinformatics and single-cell RNA sequencing analysis. This paper reviews recent developments regarding lung macrophage classification and summarizes the origin and replenishment of interstitial macrophages and their function in pulmonary fibrosis.
Collapse
Affiliation(s)
- Yanrong Gu
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Lung Stem Cells and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, Kepala Batas, Malaysia
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Toby Lawrence
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Centre for Inflammation Biology and Cancer Immunology, Cancer Research UK King’s Health Partners Centre, School of Immunology and Microbial Sciences, King’s College London, London, United Kingdom
| | - Rafeezul Mohamed
- Lung Stem Cells and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, Kepala Batas, Malaysia
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Yinming Liang, ; Badrul Hisham Yahaya,
| | - Badrul Hisham Yahaya
- Lung Stem Cells and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (AMDI), Universiti Sains Malaysia, Bertam, Kepala Batas, Malaysia
- *Correspondence: Yinming Liang, ; Badrul Hisham Yahaya,
| |
Collapse
|
22
|
Resident cardiac macrophages: Heterogeneity and function in health and disease. Immunity 2022; 55:1549-1563. [PMID: 36103852 DOI: 10.1016/j.immuni.2022.08.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 12/20/2022]
Abstract
Understanding tissue macrophage biology has become challenging in recent years due the ever-increasing complexity in macrophage-subset identification and functional characterization. This is particularly important within the myocardium, as we have come to understand that macrophages play multifaceted roles in cardiac health and disease, and heart disease remains the leading cause of death worldwide. Here, we review recent progress in the field, focusing on resident cardiac macrophage heterogeneity, origins, and functions at steady state and after injury. We stratify resident cardiac macrophage functions by the ability of macrophages to either directly influence cardiac physiology or indirectly influence cardiac physiology through orchestrating multi-cellular communication with cardiomyocytes and stromal and immune populations.
Collapse
|
23
|
Aegerter H, Lambrecht BN, Jakubzick CV. Biology of lung macrophages in health and disease. Immunity 2022; 55:1564-1580. [PMID: 36103853 DOI: 10.1016/j.immuni.2022.08.010] [Citation(s) in RCA: 187] [Impact Index Per Article: 93.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 08/08/2022] [Accepted: 08/16/2022] [Indexed: 12/14/2022]
Abstract
Tissue-resident alveolar and interstitial macrophages and recruited macrophages are critical players in innate immunity and maintenance of lung homeostasis. Until recently, assessing the differential functional contributions of tissue-resident versus recruited macrophages has been challenging because they share overlapping cell surface markers, making it difficult to separate them using conventional methods. This review describes how scRNA-seq and spatial transcriptomics can separate these subpopulations and help unravel the complexity of macrophage biology in homeostasis and disease. First, we provide a guide to identifying and distinguishing lung macrophages from other mononuclear phagocytes in humans and mice. Second, we outline emerging concepts related to the development and function of the various lung macrophages in the alveolar, perivascular, and interstitial niches. Finally, we describe how different tissue states profoundly alter their functions, including acute and chronic lung disease, cancer, and aging.
Collapse
Affiliation(s)
- Helena Aegerter
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, ErasmusMC, Rotterdam, the Netherlands
| | - Claudia V Jakubzick
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, NH, USA.
| |
Collapse
|
24
|
A cardioimmunologist's toolkit: genetic tools to dissect immune cells in cardiac disease. Nat Rev Cardiol 2022; 19:395-413. [PMID: 35523863 DOI: 10.1038/s41569-022-00701-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2022] [Indexed: 02/06/2023]
Abstract
Cardioimmunology is a field that encompasses the immune cells and pathways that modulate cardiac function in homeostasis and regulate the temporal balance between tissue injury and repair in disease. Over the past two decades, genetic fate mapping and high-dimensional sequencing techniques have defined increasing functional heterogeneity of innate and adaptive immune cell populations in the heart and other organs, revealing a complexity not previously appreciated and challenging established frameworks for the immune system. Given these rapid advances, understanding how to use these tools has become crucial. However, cardiovascular biologists without immunological expertise might not be aware of the strengths and caveats of immune-related tools and how they can be applied to examine the pathogenesis of myocardial diseases. In this Review, we guide readers through case-based examples to demonstrate how tool selection can affect data quality and interpretation and we provide critical analysis of the experimental tools that are currently available, focusing on their use in models of ischaemic heart injury and heart failure. The goal is to increase the use of relevant immunological tools and strategies among cardiovascular researchers to improve the precision, translatability and consistency of future studies of immune cells in cardiac disease.
Collapse
|
25
|
Sanin DE, Ge Y, Marinkovic E, Kabat AM, Castoldi A, Caputa G, Grzes KM, Curtis JD, Thompson EA, Willenborg S, Dichtl S, Reinhardt S, Dahl A, Pearce EL, Eming SA, Gerbaulet A, Roers A, Murray PJ, Pearce EJ. A common framework of monocyte-derived macrophage activation. Sci Immunol 2022; 7:eabl7482. [PMID: 35427180 DOI: 10.1126/sciimmunol.abl7482] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Macrophages populate every organ during homeostasis and disease, displaying features of tissue imprinting and heterogeneous activation. The disconnected picture of macrophage biology that has emerged from these observations is a barrier for integration across models or with in vitro macrophage activation paradigms. We set out to contextualize macrophage heterogeneity across mouse tissues and inflammatory conditions, specifically aiming to define a common framework of macrophage activation. We built a predictive model with which we mapped the activation of macrophages across 12 tissues and 25 biological conditions, finding a notable commonality and finite number of transcriptional profiles, in particular among infiltrating macrophages, which we modeled as defined stages along four conserved activation paths. These activation paths include a "phagocytic" regulatory path, an "inflammatory" cytokine-producing path, an "oxidative stress" antimicrobial path, or a "remodeling" extracellular matrix deposition path. We verified this model with adoptive cell transfer experiments and identified transient RELMɑ expression as a feature of monocyte-derived macrophage tissue engraftment. We propose that this integrative approach of macrophage classification allows the establishment of a common predictive framework of monocyte-derived macrophage activation in inflammation and homeostasis.
Collapse
Affiliation(s)
- David E Sanin
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.,Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yan Ge
- Institute for Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Emilija Marinkovic
- Institute for Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Agnieszka M Kabat
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.,Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Angela Castoldi
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - George Caputa
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Katarzyna M Grzes
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.,Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jonathan D Curtis
- Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth A Thompson
- Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sebastian Willenborg
- Department of Dermatology, University of Cologne, Kerpenerstr. 62, 50937 Cologne, Germany
| | - Stefanie Dichtl
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Susanne Reinhardt
- DRESDEN-concept Genome Center, TU Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Andreas Dahl
- DRESDEN-concept Genome Center, TU Dresden, Fetscherstr. 105, 01307 Dresden, Germany
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.,Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Sabine A Eming
- Department of Dermatology, University of Cologne, Kerpenerstr. 62, 50937 Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Institute of Zoology, Developmental Biology Unit, University of Cologne, Cologne, Germany
| | - Alexander Gerbaulet
- Institute for Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Axel Roers
- Institute for Immunology, Medical Faculty Carl Gustav Carus, TU Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Peter J Murray
- Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - Edward J Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.,Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany.,Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| |
Collapse
|
26
|
Honda A, Hoeksema MA, Sakai M, Lund SJ, Lakhdari O, Butcher LD, Rambaldo TC, Sekiya NM, Nasamran CA, Fisch KM, Sajti E, Glass CK, Prince LS. The Lung Microenvironment Instructs Gene Transcription in Neonatal and Adult Alveolar Macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1947-1959. [PMID: 35354612 PMCID: PMC9012679 DOI: 10.4049/jimmunol.2101192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/10/2022] [Indexed: 12/13/2022]
Abstract
Immaturity of alveolar macrophages (AMs) around birth contributes to the susceptibility of newborns to lung disease. However, the molecular features differentiating neonatal and mature, adult AMs are poorly understood. In this study, we identify the unique transcriptomes and enhancer landscapes of neonatal and adult AMs in mice. Although the core AM signature was similar, murine adult AMs expressed higher levels of genes involved in lipid metabolism, whereas neonatal AMs expressed a largely proinflammatory gene profile. Open enhancer regions identified by an assay for transposase-accessible chromatin followed by high-throughput sequencing (ATAC-seq) contained motifs for nuclear receptors, MITF, and STAT in adult AMs and AP-1 and NF-κB in neonatal AMs. Intranasal LPS activated a similar innate immune response in both neonatal and adult mice, with higher basal expression of inflammatory genes in neonates. The lung microenvironment drove many of the distinguishing gene expression and open chromatin characteristics of neonatal and adult AMs. Neonatal mouse AMs retained high expression of some proinflammatory genes, suggesting that the differences in neonatal AMs result from both inherent cell properties and environmental influences.
Collapse
Affiliation(s)
- Asami Honda
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Rady Children's Hospital, San Diego, CA
| | - Marten A Hoeksema
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Mashito Sakai
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
| | - Sean J Lund
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Rady Children's Hospital, San Diego, CA
| | - Omar Lakhdari
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Rady Children's Hospital, San Diego, CA
| | - Lindsay D Butcher
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | | | | | - Chanond A Nasamran
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA
| | - Kathleen M Fisch
- Center for Computational Biology and Bioinformatics, University of California, San Diego, La Jolla, CA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Diego, La Jolla, CA; and
| | - Eniko Sajti
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Rady Children's Hospital, San Diego, CA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Lawrence S Prince
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA;
| |
Collapse
|
27
|
Lung type II alveolar epithelial cells collaborate with CCR2+ inflammatory monocytes in host defense against poxvirus infection. Nat Commun 2022; 13:1671. [PMID: 35351885 PMCID: PMC8964745 DOI: 10.1038/s41467-022-29308-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
The pulmonary immune system consists of a network of tissue-resident cells as well as immune cells that are recruited to the lungs during infection and/or inflammation. How these immune components function during an acute poxvirus infection is not well understood. Intranasal infection of mice with vaccinia virus causes lethal pneumonia and systemic dissemination. Here we report that vaccinia C7 is a crucial virulence factor that blocks activation of the transcription factor IRF3. We provide evidence that type II alveolar epithelial cells (AECIIs) respond to pulmonary infection of vaccinia virus by inducing IFN-β and IFN-stimulated genes via the activation of the MDA5 and STING-mediated nucleic acid-sensing pathways and the type I IFN positive feedback loop. This leads to the recruitment and activation of CCR2+ inflammatory monocytes in the infected lungs and subsequent differentiation into Lyve1− interstitial macrophages (Lyve1− IMs), which efficiently engulf viral particles and block viral replication. Our results provide insights into how innate immune sensing of viral infection by lung AECIIs influences the activation and differentiation of CCR2+ inflammatory monocytes to defend against pulmonary poxvirus infection. Smallpox is a highly contagious respiratory pathogen associated with a high mortality rate. Here the authors utilize a mouse model of intranasal vaccinia virus infection and show a C7 gene encoded virulence factor attenuates type I IFN release by lung type II alveolar epithelial cells and reduces lung inflammatory monocyte responses.
Collapse
|
28
|
Abstract
Tuberculosis (TB) remains the leading cause of bacterial disease-related death and is among the top 10 overall causes of death worldwide. The complex nature of this infectious lung disease has proven difficult to treat, and significant research efforts are now evaluating the feasibility of host-directed, adjunctive therapies. An attractive approach in host-directed therapy targets host epigenetics, or gene regulation, to redirect the immune response in a host-beneficial manner. Substantial evidence exists demonstrating that host epigenetics are dysregulated during TB and that epigenetic-based therapies may be highly effective to treat TB. However, the caveat is that much of the knowledge that exists on the modulation of the host epigenome during TB has been gained using in vitro, small-animal, or blood-derived cell models, which do not accurately reflect the pulmonary nature of the disease. In humans, the first and major target cells of Mycobacterium tuberculosis are alveolar macrophages (AM). As such, their response to infection and treatment is clinically relevant and ultimately drives the outcome of disease. In this review, we compare the fundamental differences between AM and circulating monocyte-derived macrophages in the context of TB and summarize the recent advances in elucidating the epigenomes of these cells, including changes to the transcriptome, DNA methylome, and chromatin architecture. We will also discuss trained immunity in AM as a new and emerging field in TB research and provide some perspectives for the translational potential of targeting host epigenetics as an alternative TB therapy.
Collapse
|
29
|
Günes Günsel G, Conlon TM, Jeridi A, Kim R, Ertüz Z, Lang NJ, Ansari M, Novikova M, Jiang D, Strunz M, Gaianova M, Hollauer C, Gabriel C, Angelidis I, Doll S, Pestoni JC, Edelmann SL, Kohlhepp MS, Guillot A, Bassler K, Van Eeckhoutte HP, Kayalar Ö, Konyalilar N, Kanashova T, Rodius S, Ballester-López C, Genes Robles CM, Smirnova N, Rehberg M, Agarwal C, Krikki I, Piavaux B, Verleden SE, Vanaudenaerde B, Königshoff M, Dittmar G, Bracke KR, Schultze JL, Watz H, Eickelberg O, Stoeger T, Burgstaller G, Tacke F, Heissmeyer V, Rinkevich Y, Bayram H, Schiller HB, Conrad M, Schneider R, Yildirim AÖ. The arginine methyltransferase PRMT7 promotes extravasation of monocytes resulting in tissue injury in COPD. Nat Commun 2022; 13:1303. [PMID: 35288557 PMCID: PMC8921220 DOI: 10.1038/s41467-022-28809-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
Extravasation of monocytes into tissue and to the site of injury is a fundamental immunological process, which requires rapid responses via post translational modifications (PTM) of proteins. Protein arginine methyltransferase 7 (PRMT7) is an epigenetic factor that has the capacity to mono-methylate histones on arginine residues. Here we show that in chronic obstructive pulmonary disease (COPD) patients, PRMT7 expression is elevated in the lung tissue and localized to the macrophages. In mouse models of COPD, lung fibrosis and skin injury, reduced expression of PRMT7 associates with decreased recruitment of monocytes to the site of injury and hence less severe symptoms. Mechanistically, activation of NF-κB/RelA in monocytes induces PRMT7 transcription and consequential mono-methylation of histones at the regulatory elements of RAP1A, which leads to increased transcription of this gene that is responsible for adhesion and migration of monocytes. Persistent monocyte-derived macrophage accumulation leads to ALOX5 over-expression and accumulation of its metabolite LTB4, which triggers expression of ACSL4 a ferroptosis promoting gene in lung epithelial cells. Conclusively, inhibition of arginine mono-methylation might offer targeted intervention in monocyte-driven inflammatory conditions that lead to extensive tissue damage if left untreated.
Collapse
Affiliation(s)
- Gizem Günes Günsel
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Thomas M Conlon
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Aicha Jeridi
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Rinho Kim
- Institute of Functional Epigenetics, Helmholtz Munich, 85764, Munich, Germany
| | - Zeynep Ertüz
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Niklas J Lang
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Meshal Ansari
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
- Institute of Computational Biology, Helmholtz Munich, 85764, Munich, Germany
| | - Mariia Novikova
- Institute of Metabolism and Cell Death, Helmholtz Munich, 85764, Munich, Germany
- Pirogov Russian National Research Medical University, Laboratory of Experimental Oncology, Ostrovityanova 1, Moscow, 117997, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Ostrovityanova1 bldg 10, 117997, Moscow, Russia
| | - Dongsheng Jiang
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Maximilian Strunz
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Mariia Gaianova
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Christine Hollauer
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Christina Gabriel
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Ilias Angelidis
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Sebastian Doll
- Institute of Computational Biology, Helmholtz Munich, 85764, Munich, Germany
| | - Jeanine C Pestoni
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Stephanie L Edelmann
- Research Unit Molecular Immune Regulation, Helmholtz Munich, 81377, Munich, Germany
| | - Marlene Sophia Kohlhepp
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), 13353, Berlin, Germany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), 13353, Berlin, Germany
| | - Kevin Bassler
- Department for Genomics & Immunoregulation, LIMES-Institute, University of Bonn, 53115, Bonn, Germany
- aimed analytics, 53121, Bonn, Germany
| | - Hannelore P Van Eeckhoutte
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University, University Hospital Ghent, 9000, Ghent, Belgium
| | - Özgecan Kayalar
- Koç University Research Center for Translational Medicine (KUTTAM), 34010, Istanbul, Turkey
| | - Nur Konyalilar
- Koç University Research Center for Translational Medicine (KUTTAM), 34010, Istanbul, Turkey
| | - Tamara Kanashova
- Max-Delbrück Center for Molecular Medicine, 13125, Berlin, Germany
| | - Sophie Rodius
- Proteomics of cellular signalling, Luxembourg Institute of Health, 1272, Strassen, Luxembourg
| | - Carolina Ballester-López
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | | | - Natalia Smirnova
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Denver, CO, 80045, USA
| | - Markus Rehberg
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Charu Agarwal
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Ioanna Krikki
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Benoit Piavaux
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, 25242, Vestec, Czech Republic
| | - Stijn E Verleden
- Division of Pneumology, KU Leuven, 3000, Leuven, Belgium
- Antwerp Surgical Training, Anatomy and Research Centre, University of Antwerp, 2650, Edegem, Belgium
| | | | - Melanie Königshoff
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Gunnar Dittmar
- Proteomics of cellular signalling, Luxembourg Institute of Health, 1272, Strassen, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Ken R Bracke
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University, University Hospital Ghent, 9000, Ghent, Belgium
| | - Joachim L Schultze
- Department for Genomics & Immunoregulation, LIMES-Institute, University of Bonn, 53115, Bonn, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) e.V., PRECISE Platform for Single Cell Genomics and Epigenomics at DZNE and the University of Bonn, 53115, Bonn, Germany
| | - Henrik Watz
- Pulmonary Research Institute at LungenClinic Grosshansdorf, Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927, Grosshansdorf, Germany
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Tobias Stoeger
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Gerald Burgstaller
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), 13353, Berlin, Germany
| | - Vigo Heissmeyer
- Research Unit Molecular Immune Regulation, Helmholtz Munich, 81377, Munich, Germany
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, 82152, Planegg-Martinsried, Germany
| | - Yuval Rinkevich
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Hasan Bayram
- Koç University Research Center for Translational Medicine (KUTTAM), 34010, Istanbul, Turkey
| | - Herbert B Schiller
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Munich, 85764, Munich, Germany
- Pirogov Russian National Research Medical University, Laboratory of Experimental Oncology, Ostrovityanova 1, Moscow, 117997, Russia
| | - Robert Schneider
- Institute of Functional Epigenetics, Helmholtz Munich, 85764, Munich, Germany
| | - Ali Önder Yildirim
- Institute of Lung Health and Immunity (LHI), Comprehensive Pneumology Center (CPC), Helmholtz Munich, Member of the German Center for Lung Research (DZL), 85764, Munich, Germany.
- Koç University Research Center for Translational Medicine (KUTTAM), 34010, Istanbul, Turkey.
| |
Collapse
|
30
|
de Macedo LH, Souza COS, Gardinassi LG, Faccioli LH. CD14 regulates the metabolomic profiles of distinct macrophage subsets under steady and activated states. Immunobiology 2022; 227:152191. [DOI: 10.1016/j.imbio.2022.152191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 01/28/2022] [Accepted: 02/14/2022] [Indexed: 11/05/2022]
|
31
|
Zhang B, Zhang Y, Xiong L, Li Y, Zhang Y, Zhao J, Jiang H, Li C, Liu Y, Liu X, Liu H, Ping YF, Zhang QC, Zhang Z, Bian XW, Zhao Y, Hu X. CD127 imprints functional heterogeneity to diversify monocyte responses in inflammatory diseases. J Exp Med 2022; 219:e20211191. [PMID: 35015026 PMCID: PMC8757045 DOI: 10.1084/jem.20211191] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/21/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory monocytes are key mediators of acute and chronic inflammation; yet, their functional diversity remains obscure. Single-cell transcriptome analyses of human inflammatory monocytes from COVID-19 and rheumatoid arthritis patients revealed a subset of cells positive for CD127, an IL-7 receptor subunit, and such positivity rendered otherwise inert monocytes responsive to IL-7. Active IL-7 signaling engaged epigenetically coupled, STAT5-coordinated transcriptional programs to restrain inflammatory gene expression, resulting in inverse correlation between CD127 expression and inflammatory phenotypes in a seemingly homogeneous monocyte population. In COVID-19 and rheumatoid arthritis, CD127 marked a subset of monocytes/macrophages that retained hypoinflammatory phenotypes within the highly inflammatory tissue environments. Furthermore, generation of an integrated expression atlas revealed unified features of human inflammatory monocytes across different diseases and different tissues, exemplified by those of the CD127high subset. Overall, we phenotypically and molecularly characterized CD127-imprinted functional heterogeneity of human inflammatory monocytes with direct relevance for inflammatory diseases.
Collapse
Affiliation(s)
- Bin Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Yuan Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Lei Xiong
- Ministry of Education Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuzhe Li
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yunliang Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Jiuliang Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunological Diseases, Beijing, China
| | - Hui Jiang
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunological Diseases, Beijing, China
| | - Can Li
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunological Diseases, Beijing, China
| | - Yunqi Liu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
| | - Xindong Liu
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Haofei Liu
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi-Fang Ping
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qiangfeng Cliff Zhang
- Ministry of Education Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, Shenzhen, China
- The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiu-Wu Bian
- Institute of Pathology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunological Diseases, Beijing, China
| | - Xiaoyu Hu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Center for Human Disease Immuno-monitoring, Beijing Friendship Hospital, Beijing, China
| |
Collapse
|
32
|
Roquilly A, Mintern JD, Villadangos JA. Spatiotemporal Adaptations of Macrophage and Dendritic Cell Development and Function. Annu Rev Immunol 2022; 40:525-557. [PMID: 35130030 DOI: 10.1146/annurev-immunol-101320-031931] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Macrophages and conventional dendritic cells (cDCs) are distributed throughout the body, maintaining tissue homeostasis and tolerance to self and orchestrating innate and adaptive immunity against infection and cancer. As they complement each other, it is important to understand how they cooperate and the mechanisms that integrate their functions. Both are exposed to commensal microbes, pathogens, and other environmental challenges that differ widely among anatomical locations and over time. To adjust to these varying conditions, macrophages and cDCs acquire spatiotemporal adaptations (STAs) at different stages of their life cycle that determine how they respond to infection. The STAs acquired in response to previous infections can result in increased responsiveness to infection, termed training, or in reduced responses, termed paralysis, which in extreme cases can cause immunosuppression. Understanding the developmental stage and location where macrophages and cDCs acquire their STAs, and the molecular and cellular players involved in their induction, may afford opportunities to harness their beneficial outcomes and avoid or reverse their deleterious effects. Here we review our current understanding of macrophage and cDC development, life cycle, function, and STA acquisition before, during, and after infection. We propose a unified framework to explain how these two cell types adjust their activities to changing conditions over space and time to coordinate their immunosurveillance functions. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Antoine Roquilly
- Center for Research in Transplantation and Translational Immunology, INSERM, UMR 1064, CHU Nantes, University of Nantes, Nantes, France
| | - Justine D Mintern
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Jose A Villadangos
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.,Department of Microbiology and Immunology, Doherty Institute of Infection and Immunity, The University of Melbourne, Parkville, Victoria, Australia;
| |
Collapse
|
33
|
Bain CC, MacDonald AS. The impact of the lung environment on macrophage development, activation and function: diversity in the face of adversity. Mucosal Immunol 2022; 15:223-234. [PMID: 35017701 PMCID: PMC8749355 DOI: 10.1038/s41385-021-00480-w] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/04/2021] [Accepted: 12/18/2021] [Indexed: 02/04/2023]
Abstract
The last decade has been somewhat of a renaissance period for the field of macrophage biology. This renewed interest, combined with the advent of new technologies and development of novel model systems to assess different facets of macrophage biology, has led to major advances in our understanding of the diverse roles macrophages play in health, inflammation, infection and repair, and the dominance of tissue environments in influencing all of these areas. Here, we discuss recent developments in our understanding of lung macrophage heterogeneity, ontogeny, metabolism and function in the context of health and disease, and highlight core conceptual advances and key unanswered questions that we believe should be focus of work in the coming years.
Collapse
Affiliation(s)
- Calum C Bain
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh Bioquarter, Edinburgh, EH16 4TJ, UK.
| | - Andrew S MacDonald
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, M13 9NT, UK.
| |
Collapse
|
34
|
Sun X, Perl AK, Li R, Bell SM, Sajti E, Kalinichenko VV, Kalin TV, Misra RS, Deshmukh H, Clair G, Kyle J, Crotty Alexander LE, Masso-Silva JA, Kitzmiller JA, Wikenheiser-Brokamp KA, Deutsch G, Guo M, Du Y, Morley MP, Valdez MJ, Yu HV, Jin K, Bardes EE, Zepp JA, Neithamer T, Basil MC, Zacharias WJ, Verheyden J, Young R, Bandyopadhyay G, Lin S, Ansong C, Adkins J, Salomonis N, Aronow BJ, Xu Y, Pryhuber G, Whitsett J, Morrisey EE. A census of the lung: CellCards from LungMAP. Dev Cell 2022; 57:112-145.e2. [PMID: 34936882 PMCID: PMC9202574 DOI: 10.1016/j.devcel.2021.11.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 07/19/2021] [Accepted: 11/05/2021] [Indexed: 01/07/2023]
Abstract
The human lung plays vital roles in respiration, host defense, and basic physiology. Recent technological advancements such as single-cell RNA sequencing and genetic lineage tracing have revealed novel cell types and enriched functional properties of existing cell types in lung. The time has come to take a new census. Initiated by members of the NHLBI-funded LungMAP Consortium and aided by experts in the lung biology community, we synthesized current data into a comprehensive and practical cellular census of the lung. Identities of cell types in the normal lung are captured in individual cell cards with delineation of function, markers, developmental lineages, heterogeneity, regenerative potential, disease links, and key experimental tools. This publication will serve as the starting point of a live, up-to-date guide for lung research at https://www.lungmap.net/cell-cards/. We hope that Lung CellCards will promote the community-wide effort to establish, maintain, and restore respiratory health.
Collapse
Affiliation(s)
- Xin Sun
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | - Anne-Karina Perl
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Rongbo Li
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Sheila M Bell
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Eniko Sajti
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Vladimir V Kalinichenko
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA; Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Tanya V Kalin
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Ravi S Misra
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Hitesh Deshmukh
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jennifer Kyle
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Laura E Crotty Alexander
- Deparment of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jorge A Masso-Silva
- Deparment of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Joseph A Kitzmiller
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Kathryn A Wikenheiser-Brokamp
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pathology & Laboratory Medicine, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Gail Deutsch
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA; Department of Laboratories, Seattle Children's Hospital, OC.8.720, 4800 Sand Point Way Northeast, Seattle, WA 98105, USA
| | - Minzhe Guo
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Yina Du
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Michael P Morley
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Valdez
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Haoze V Yu
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kang Jin
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Eric E Bardes
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jarod A Zepp
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Terren Neithamer
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria C Basil
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - William J Zacharias
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Internal Medicine, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Jamie Verheyden
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Randee Young
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Gautam Bandyopadhyay
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sara Lin
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles Ansong
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Joshua Adkins
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Nathan Salomonis
- Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Bruce J Aronow
- Departments of Biomedical Informatics, Developmental Biology, and Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Yan Xu
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Gloria Pryhuber
- Department of Pediatrics Division of Neonatology, The University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jeff Whitsett
- Division of Neonatology and Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3230 Eden Avenue, Cincinnati, OH 45267, USA
| | - Edward E Morrisey
- Penn-CHOP Lung Biology Institute, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Dick SA, Wong A, Hamidzada H, Nejat S, Nechanitzky R, Vohra S, Mueller B, Zaman R, Kantores C, Aronoff L, Momen A, Nechanitzky D, Li WY, Ramachandran P, Crome SQ, Becher B, Cybulsky MI, Billia F, Keshavjee S, Mital S, Robbins CS, Mak TW, Epelman S. Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles. Sci Immunol 2022; 7:eabf7777. [PMID: 34995099 DOI: 10.1126/sciimmunol.abf7777] [Citation(s) in RCA: 192] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Sarah A Dick
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada
| | - Anthony Wong
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Homaira Hamidzada
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sara Nejat
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada
| | - Robert Nechanitzky
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, ON, Canada
| | - Shabana Vohra
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada.,Peter Munk Cardiac Centre, UHN, Toronto, ON, Canada
| | | | - Rysa Zaman
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Crystal Kantores
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada
| | - Laura Aronoff
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Abdul Momen
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada
| | - Duygu Nechanitzky
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, ON, Canada
| | - Wanda Y Li
- Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, ON, Canada
| | | | - Sarah Q Crome
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich, Zürich 8057, Switzerland
| | - Myron I Cybulsky
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada.,Peter Munk Cardiac Centre, UHN, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Filio Billia
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada.,Peter Munk Cardiac Centre, UHN, Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Depatment of Pathology, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Shaf Keshavjee
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Toronto Lung Transplant Program, UHN Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Seema Mital
- Division of Cardiology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Clint S Robbins
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada.,Peter Munk Cardiac Centre, UHN, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tak W Mak
- Department of Immunology, University of Toronto, Toronto, ON, Canada.,Princess Margaret Cancer Centre, University Health Network (UHN), Toronto, ON, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.,Depatment of Pathology, University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network (UHN), Toronto, ON, Canada.,Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada.,Peter Munk Cardiac Centre, UHN, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
36
|
McCowan J, Fercoq F, Kirkwood PM, T’Jonck W, Hegarty LM, Mawer CM, Cunningham R, Mirchandani AS, Hoy A, Humphries DC, Jones GR, Hansen CG, Hirani N, Jenkins SJ, Henri S, Malissen B, Walmsley SR, Dockrell DH, Saunders PTK, Carlin LM, Bain CC. The transcription factor EGR2 is indispensable for tissue-specific imprinting of alveolar macrophages in health and tissue repair. Sci Immunol 2021; 6:eabj2132. [PMID: 34797692 PMCID: PMC7612216 DOI: 10.1126/sciimmunol.abj2132] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Alveolar macrophages are the most abundant macrophages in the healthy lung where they play key roles in homeostasis and immune surveillance against airborne pathogens. Tissue-specific differentiation and survival of alveolar macrophages rely on niche-derived factors, such as granulocyte-macrophage colony-stimulating factor (GM-CSF) and transforming growth factor–β (TGF-β). However, the nature of the downstream molecular pathways that regulate the identity and function of alveolar macrophages and their response to injury remain poorly understood. Here, we identify that the transcription factor EGR2 is an evolutionarily conserved feature of lung alveolar macrophages and show that cell-intrinsic EGR2 is indispensable for the tissue-specific identity of alveolar macrophages. Mechanistically, we show that EGR2 is driven by TGF-β and GM-CSF in a PPAR-γ–dependent manner to control alveolar macrophage differentiation. Functionally, EGR2 was dispensable for the regulation of lipids in the airways but crucial for the effective handling of the respiratory pathogen Streptococcus pneumoniae. Last, we show that EGR2 is required for repopulation of the alveolar niche after sterile, bleomycin-induced lung injury and demonstrate that EGR2-dependent, monocyte-derived alveolar macrophages are vital for effective tissue repair after injury. Collectively, we demonstrate that EGR2 is an indispensable component of the transcriptional network controlling the identity and function of alveolar macrophages in health and disease.
Collapse
Affiliation(s)
- Jack McCowan
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | | | - Phoebe M. Kirkwood
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Wouter T’Jonck
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Lizi M. Hegarty
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Connar M. Mawer
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
| | - Richard Cunningham
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Ananda S. Mirchandani
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Anna Hoy
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
| | - Duncan C. Humphries
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Gareth-Rhys Jones
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Carsten G. Hansen
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Nik Hirani
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Stephen J. Jenkins
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Sandrine Henri
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, INSERM, U1104, CNRS UMR7280, 13288 Marseille, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, INSERM, U1104, CNRS UMR7280, 13288 Marseille, France
| | - Sarah R. Walmsley
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - David H. Dockrell
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Philippa T. K. Saunders
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| | - Leo M. Carlin
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Calum C. Bain
- University of Edinburgh Centre for Inflammation Research, Queens Medical Research Institute, 47 Little France Crescent, Edinburgh BioQuarter, Edinburgh, EH16 4TJ, UK
- Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, UK
| |
Collapse
|
37
|
Hou F, Xiao K, Tang L, Xie L. Diversity of Macrophages in Lung Homeostasis and Diseases. Front Immunol 2021; 12:753940. [PMID: 34630433 PMCID: PMC8500393 DOI: 10.3389/fimmu.2021.753940] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/08/2021] [Indexed: 01/14/2023] Open
Abstract
Lung macrophages play important roles in the maintenance of homeostasis, pathogen clearance and immune regulation. The different types of pulmonary macrophages and their roles in lung diseases have attracted attention in recent years. Alveolar macrophages (AMs), including tissue-resident alveolar macrophages (TR-AMs) and monocyte-derived alveolar macrophages (Mo-AMs), as well as interstitial macrophages (IMs) are the major macrophage populations in the lung and have unique characteristics in both steady-state conditions and disease states. The different characteristics of these three types of macrophages determine the different roles they play in the development of disease. Therefore, it is important to fully understand the similarities and differences among these three types of macrophages for the study of lung diseases. In this review, we will discuss the physiological characteristics and unique functions of these three types of macrophages in acute and chronic lung diseases. We will also discuss possible methods to target macrophages in lung diseases.
Collapse
Affiliation(s)
- Fei Hou
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China.,Medical School of Chinese PLA, Beijing, China
| | - Kun Xiao
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| | - Li Tang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences·Beijing, Beijing Institute of Lifeomics, Beijing, China
| | - Lixin Xie
- College of Pulmonary and Critical Care Medicine, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
38
|
Troutman TD, Kofman E, Glass CK. Exploiting dynamic enhancer landscapes to decode macrophage and microglia phenotypes in health and disease. Mol Cell 2021; 81:3888-3903. [PMID: 34464593 PMCID: PMC8500948 DOI: 10.1016/j.molcel.2021.08.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/19/2021] [Accepted: 07/30/2021] [Indexed: 12/24/2022]
Abstract
The development and functional potential of metazoan cells is dependent on combinatorial roles of transcriptional enhancers and promoters. Macrophages provide exceptionally powerful model systems for investigation of mechanisms underlying the activation of cell-specific enhancers that drive transitions in cell fate and cell state. Here, we review recent advances that have expanded appreciation of the diversity of macrophage phenotypes in health and disease, emphasizing studies of liver, adipose tissue, and brain macrophages as paradigms for other tissue macrophages and cell types. Studies of normal tissue-resident macrophages and macrophages associated with cirrhosis, obese adipose tissue, and neurodegenerative disease illustrate the major roles of tissue environment in remodeling enhancer landscapes to specify the development and functions of distinct macrophage phenotypes. We discuss the utility of quantitative analysis of environment-dependent changes in enhancer activity states as an approach to discovery of regulatory transcription factors and upstream signaling pathways.
Collapse
Affiliation(s)
- Ty D Troutman
- Department of Medicine, University of California, San Diego, San Diego, CA, USA; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Eric Kofman
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA; Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, San Diego, CA, USA
| | - Christopher K Glass
- Department of Medicine, University of California, San Diego, San Diego, CA, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
39
|
Alobaidi A, Alsamarai A, Alsamarai MA. Inflammation in Asthma Pathogenesis: Role of T cells, Macrophages, Epithelial Cells and Type 2 Inflammation. Antiinflamm Antiallergy Agents Med Chem 2021; 20:317-332. [PMID: 34544350 DOI: 10.2174/1871523020666210920100707] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/06/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
Asthma is a chronic disease with abnormal inflammatory and immunological responses. The disease initiated by antigens in subjects with genetic susceptibility. However, environmental factors play a role in the initiation and exacerbation of asthma attack. Asthma is T helper 2 (Th2)-cell-mediated disease. Recent studies indicated that asthma is not a single disease entity, but it is with multiple phenotypes and endotypes. The pathophysiological changes in asthma included a series of subsequent continuous vicious circle of cellular activation contributed to induction of chemokines and cytokines that potentiate inflammation. The heterogeneity of asthma influenced the treatment response. The asthma pathogenesis driven by varied set of cells such as eosinophils, basophils, neutrophils, mast cells, macrophages, epithelial cells and T cells. In this review the role of T cells, macrophage, and epithelial cells are discussed.
Collapse
Affiliation(s)
- Amina Alobaidi
- Kirkuk University College of Veterinary Medicine, Kirkuk. Iraq
| | - Abdulghani Alsamarai
- Aalborg Academy College of Medicine [AACOM], Denmark. Tikrit University College of Medicine, [TUCOM], Tikrit. Iraq
| | | |
Collapse
|
40
|
Huggins DN, LaRue RS, Wang Y, Knutson TP, Xu Y, Williams JW, Schwertfeger KL. Characterizing macrophage diversity in metastasis-bearing lungs reveals a lipid-associated macrophage subset. Cancer Res 2021; 81:5284-5295. [PMID: 34389631 DOI: 10.1158/0008-5472.can-21-0101] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 07/15/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022]
Abstract
While macrophages are among the most abundant immune cell type found within primary and metastatic mammary tumors, how their complexity and heterogeneity change with metastatic progression remains unknown. Here, macrophages were isolated from the lungs of mice bearing orthotopic mammary tumors for single-cell RNA sequencing. Seven distinct macrophage clusters were identified, including populations exhibiting enhanced differential expression of genes related to antigen presentation (H2-Aa, Cd74), cell cycle (Stmn1, Cdk1), and interferon signaling (Isg15, Ifitm3). Interestingly, one cluster demonstrated a profile concordant with lipid-associated macrophages (Lgals3, Trem2). Compared to non-tumor-bearing controls, the number of these cells per gram of tissue was significantly increased in lungs from tumor-bearing mice, with the vast majority co-staining positively with the alveolar macrophage marker Siglec-F. Enrichment of genes implicated in pathways related to lipid metabolism as well extracellular matrix remodeling and immunosuppression was observed. Additionally, these cells displayed reduced capacity for phagocytosis. Collectively, these findings highlight the diversity of macrophages present within metastatic lesions and characterize a lipid-associated macrophage subset previously unidentified in lung metastases.
Collapse
Affiliation(s)
- Danielle N Huggins
- Department of Laboratory Medicine and Pathology, University of Minnesota
| | - Rebecca S LaRue
- Genetics, Cell Biology and Development, University of Minnesota
| | - Ying Wang
- Masonic Cancer Center, University of Minnesota
| | | | | | | | | |
Collapse
|
41
|
Rasheed A, Rayner KJ. Macrophage Responses to Environmental Stimuli During Homeostasis and Disease. Endocr Rev 2021; 42:407-435. [PMID: 33523133 PMCID: PMC8284619 DOI: 10.1210/endrev/bnab004] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/20/2022]
Abstract
Work over the last 40 years has described macrophages as a heterogeneous population that serve as the frontline surveyors of tissue immunity. As a class, macrophages are found in almost every tissue in the body and as distinct populations within discrete microenvironments in any given tissue. During homeostasis, macrophages protect these tissues by clearing invading foreign bodies and/or mounting immune responses. In addition to varying identities regulated by transcriptional programs shaped by their respective environments, macrophage metabolism serves as an additional regulator to temper responses to extracellular stimuli. The area of research known as "immunometabolism" has been established within the last decade, owing to an increase in studies focusing on the crosstalk between altered metabolism and the regulation of cellular immune processes. From this research, macrophages have emerged as a prime focus of immunometabolic studies, although macrophage metabolism and their immune responses have been studied for centuries. During disease, the metabolic profile of the tissue and/or systemic regulators, such as endocrine factors, become increasingly dysregulated. Owing to these changes, macrophage responses can become skewed to promote further pathophysiologic changes. For instance, during diabetes, obesity, and atherosclerosis, macrophages favor a proinflammatory phenotype; whereas in the tumor microenvironment, macrophages elicit an anti-inflammatory response to enhance tumor growth. Herein we have described how macrophages respond to extracellular cues including inflammatory stimuli, nutrient availability, and endocrine factors that occur during and further promote disease progression.
Collapse
Affiliation(s)
- Adil Rasheed
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Katey J Rayner
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
42
|
Soysa R, Bean JC, Wu X, Lampert S, Yuen S, Crispe IN. Early-Derived Murine Macrophages Temporarily Renounce Tissue Identity during Acute Systemic Inflammation. THE JOURNAL OF IMMUNOLOGY 2021; 207:569-576. [PMID: 34193604 DOI: 10.4049/jimmunol.2001324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 05/09/2021] [Indexed: 12/24/2022]
Abstract
In mice, a subset of cardiac macrophages and Kupffer cells derive from fetal precursors, seed the developing tissues, self-renew locally, and persist into adulthood. In this study we investigated how these cells survive acute systemic inflammation. In both tissues, early-derived subsets rapidly responded to acute systemic inflammation by assuming a temporary nonclassical activation state featuring upregulation of both proinflammatory (Il1b, Tnf, Nfkb1), and anti-inflammatory (Il10, Il4ra, Nfkbiz) genes. During this process, transcription factor genes associated with myeloid identity (Spi1, Zeb2) were upregulated, whereas those associated with tissue specificity (Nr1h3 for Kupffer cells and Nfatc2 and Irf4 for cardiac macrophages) were downregulated, suggesting that the cells reasserted their myeloid identity but renounced their tissue identity. Most of these changes in gene expression reverted to steady-state levels postresolution. We conclude that these early-derived macrophage subsets are resilient in the face of acute stress by temporary loss of adaptation to local tissue-specific niches while reasserting their generic myeloid identity.
Collapse
Affiliation(s)
- Radika Soysa
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, WA; and
| | | | - Xia Wu
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, WA; and
| | - Sarah Lampert
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, WA; and
| | - Sebastian Yuen
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, WA; and
| | - Ian N Crispe
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, WA; and
| |
Collapse
|
43
|
Mould KJ, Moore CM, McManus SA, McCubbrey AL, McClendon JD, Griesmer CL, Henson PM, Janssen WJ. Airspace Macrophages and Monocytes Exist in Transcriptionally Distinct Subsets in Healthy Adults. Am J Respir Crit Care Med 2021; 203:946-956. [PMID: 33079572 PMCID: PMC8048748 DOI: 10.1164/rccm.202005-1989oc] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/15/2020] [Indexed: 01/06/2023] Open
Abstract
Rationale: Macrophages are the most abundant immune cell in the alveoli and small airways and are traditionally viewed as a homogeneous population during health. Whether distinct subsets of airspace macrophages are present in healthy humans is unknown. Single-cell RNA sequencing allows for examination of transcriptional heterogeneity between cells and between individuals. Understanding the conserved repertoire of airspace macrophages during health is essential to understanding cellular programing during disease.Objectives: We sought to determine the transcriptional heterogeneity of human cells obtained from BAL of healthy adults.Methods: Ten subjects underwent bronchoscopy with BAL. Cells from lavage were subjected to single-cell RNA sequencing. Unique cell populations and putative functions were identified. Transcriptional profiles were compared across individuals.Measurements and Main Results: We identify two novel subgroups of resident airspace macrophages-defined by proinflammatory and metallothionein gene expression profiles. We define subsets of monocyte-like cells and compare them with peripheral blood mononuclear cells. Finally, we compare global macrophage and monocyte programing between males and females.Conclusions: Healthy human airspaces contain multiple populations of myeloid cells that are highly conserved between individuals and between sexes. Resident macrophages make up the largest population and include novel subsets defined by inflammatory and metal-binding profiles. Monocyte-like cells within the airspaces are transcriptionally aligned with circulating blood cells and include a rare population defined by expression of cell-matrix interaction genes. This study is the first to delineate the conserved heterogeneity of airspace immune cells during health and identifies two previously unrecognized macrophage subsets.
Collapse
Affiliation(s)
- Kara J. Mould
- Department of Medicine
- Department of Biomedical Research, and
| | - Camille M. Moore
- Department of Pediatrics, National Jewish Health, Denver, Colorado
- Department of Medicine, University of Colorado, Aurora, Colorado; and
| | | | | | | | | | - Peter M. Henson
- Department of Biomedical Research, and
- Department of Biostatistics and Informatics, University of Colorado, Denver, Colorado
| | | |
Collapse
|
44
|
Does tissue imprinting restrict macrophage plasticity? Nat Immunol 2021; 22:118-127. [PMID: 33462453 DOI: 10.1038/s41590-020-00849-2] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 11/20/2020] [Indexed: 01/29/2023]
Abstract
Macrophages have long been considered as particularly plastic cells. However, recent work combining fate mapping, single-cell transcriptomics and epigenetics has undermined the macrophage plasticity dogma. Here, we discuss recent studies that have carefully dissected the response of individual macrophage subsets to pulmonary insults and call for an adjustment of the macrophage plasticity concept. We hypothesize that prolonged tissue residency shuts down much of the plasticity of macrophages and propose that the restricted plasticity of resident macrophages has been favored by evolution to safeguard tissue homeostasis. Recruited monocytes are more plastic and their differentiation into resident macrophages during inflammation can result in a dual imprinting from both the ongoing inflammation and the macrophage niche. This results in inflammation-imprinted resident macrophages, and we speculate that rewired niche circuits could maintain this inflammatory state. We believe that this revisited plasticity model offers opportunities to reset the macrophage pool after a severe inflammatory episode.
Collapse
|
45
|
Evren E, Ringqvist E, Tripathi KP, Sleiers N, Rives IC, Alisjahbana A, Gao Y, Sarhan D, Halle T, Sorini C, Lepzien R, Marquardt N, Michaëlsson J, Smed-Sörensen A, Botling J, Karlsson MCI, Villablanca EJ, Willinger T. Distinct developmental pathways from blood monocytes generate human lung macrophage diversity. Immunity 2020; 54:259-275.e7. [PMID: 33382972 DOI: 10.1016/j.immuni.2020.12.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/15/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
The study of human macrophages and their ontogeny is an important unresolved issue. Here, we use a humanized mouse model expressing human cytokines to dissect the development of lung macrophages from human hematopoiesis in vivo. Human CD34+ hematopoietic stem and progenitor cells (HSPCs) generated three macrophage populations, occupying separate anatomical niches in the lung. Intravascular cell labeling, cell transplantation, and fate-mapping studies established that classical CD14+ blood monocytes derived from HSPCs migrated into lung tissue and gave rise to human interstitial and alveolar macrophages. In contrast, non-classical CD16+ blood monocytes preferentially generated macrophages resident in the lung vasculature (pulmonary intravascular macrophages). Finally, single-cell RNA sequencing defined intermediate differentiation stages in human lung macrophage development from blood monocytes. This study identifies distinct developmental pathways from circulating monocytes to lung macrophages and reveals how cellular origin contributes to human macrophage identity, diversity, and localization in vivo.
Collapse
Affiliation(s)
- Elza Evren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Emma Ringqvist
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Kumar Parijat Tripathi
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Natalie Sleiers
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Inés Có Rives
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Arlisa Alisjahbana
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Yu Gao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Dhifaf Sarhan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Tor Halle
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Chiara Sorini
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Rico Lepzien
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Nicole Marquardt
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Jakob Michaëlsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 171 64 Stockholm, Sweden
| | - Eduardo J Villablanca
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Tim Willinger
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, 141 52 Stockholm, Sweden.
| |
Collapse
|
46
|
Kwak T, Wang F, Deng H, Condamine T, Kumar V, Perego M, Kossenkov A, Montaner LJ, Xu X, Xu W, Zheng C, Schuchter LM, Amaravadi RK, Mitchell TC, Karakousis GC, Mulligan C, Nam B, Masters G, Hockstein N, Bennett J, Nefedova Y, Gabrilovich DI. Distinct Populations of Immune-Suppressive Macrophages Differentiate from Monocytic Myeloid-Derived Suppressor Cells in Cancer. Cell Rep 2020; 33:108571. [PMID: 33378668 PMCID: PMC7809772 DOI: 10.1016/j.celrep.2020.108571] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 10/25/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022] Open
Abstract
Here, we report that functional heterogeneity of macrophages in cancer could be determined by the nature of their precursors: monocytes (Mons) and monocytic myeloid-derived suppressor cells (M-MDSCs). Macrophages that are differentiated from M-MDSCs, but not from Mons, are immune suppressive, with a genomic profile matching that of M-MDSCs. Immune-suppressive activity of M-MDSC-derived macrophages is dependent on the persistent expression of S100A9 protein in these cells. S100A9 also promotes M2 polarization of macrophages. Tissue-resident- and Mon-derived macrophages lack expression of this protein. S100A9-dependent immune-suppressive activity of macrophages involves transcription factor C/EBPβ. The presence of S100A9-positive macrophages in tumor tissues is associated with shorter survival in patients with head and neck cancer and poor response to PD-1 antibody treatment in patients with metastatic melanoma. Thus, this study reveals the pathway of the development of immune-suppressive macrophages and suggests an approach to their selective targeting.
Collapse
Affiliation(s)
| | - Fang Wang
- The Wistar Institute, Philadelphia, PA 19104, USA
| | - Hui Deng
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | - Vinit Kumar
- The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | | - Xiaowei Xu
- Tara Miller Melanoma Center, Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wei Xu
- Tara Miller Melanoma Center, Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cathy Zheng
- Tara Miller Melanoma Center, Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lynn M Schuchter
- Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi K Amaravadi
- Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara C Mitchell
- Abramson Cancer Center and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgos C Karakousis
- Abramson Cancer Center and Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Charles Mulligan
- Helen F Graham Cancer Center and Research Institute, Christiana Care, Newark, DE 19713, USA
| | - Brian Nam
- Helen F Graham Cancer Center and Research Institute, Christiana Care, Newark, DE 19713, USA
| | - Gregory Masters
- Helen F Graham Cancer Center and Research Institute, Christiana Care, Newark, DE 19713, USA
| | - Neil Hockstein
- Helen F Graham Cancer Center and Research Institute, Christiana Care, Newark, DE 19713, USA
| | - Joseph Bennett
- Helen F Graham Cancer Center and Research Institute, Christiana Care, Newark, DE 19713, USA
| | | | | |
Collapse
|
47
|
Trzebanski S, Jung S. Plasticity of monocyte development and monocyte fates. Immunol Lett 2020; 227:66-78. [PMID: 32814154 DOI: 10.1016/j.imlet.2020.07.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 01/01/2023]
Abstract
Monocytes are circulating myeloid immune precursor cells that are generated in the bone marrow. Mature monocytes are released into the circulation and, in case of need, recruited to peripheral sites of inflammation to differentiate into monocyte-derived effector cells. In absence of overt inflammation, monocytes also extravasate into selected tissues, where they complement tissue-resident macrophage compartments. Adjustment of these homeostatic monocyte infiltrates to local environment is critical to maintain health, as best established for the intestine. Defined gene expression changes that differ between gut segments presumably help strike the fine balance between the crucial function of these monocyte-derived macrophages as tissue rheostats and their detrimental hyperactivation. Environmental factors that dictate local monocyte differentiation remain incompletely understood. Definition of the latter could aid our general understanding of in vivo monocyte functions and their relation to inflammatory disorders. In this review, we summarize recent advances in our understanding of monocyte subsets, their differentiation into tissue macrophages, and selected contributions of monocyte-derived cells to steady-state physiology. Moreover, we will discuss emerging evidence for an intriguing bifurcation of monocyte development in the bone marrow and potential functional implications. Emphasis will be given to points of controversies, but we will largely focus on the healthy organism. For a discussion of monocyte and macrophage contributions to inflammatory conditions, we refer the reader to other dedicated reviews.
Collapse
Affiliation(s)
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
48
|
Kulikauskaite J, Wack A. Teaching Old Dogs New Tricks? The Plasticity of Lung Alveolar Macrophage Subsets. Trends Immunol 2020; 41:864-877. [PMID: 32896485 PMCID: PMC7472979 DOI: 10.1016/j.it.2020.08.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/24/2022]
Abstract
Alveolar macrophages (AMs) are highly abundant lung cells with important roles in homeostasis and immunity. Their function influences the outcome of lung infections, lung cancer, and chronic inflammatory disease. Recent findings reveal functional heterogeneity of AMs. Following lung insult, resident AMs can either remain unchanged, acquire new functionality, or be replaced by monocyte-derived AMs. Evidence from mouse models correlates AM function with their embryonic or monocyte origin. We hypothesize that resident AMs are terminally differentiated cells with low responsiveness and limited plasticity, while recruited, monocyte-derived AMs are initially highly immunoreactive but more plastic, able to change their function in response to environmental cues. Understanding cell-intrinsic and -extrinsic mechanisms determining AM function may provide opportunities for intervention in lung disease.
Collapse
Affiliation(s)
| | - Andreas Wack
- Immunoregulation Laboratory, Francis Crick Institute, London, UK.
| |
Collapse
|
49
|
The Role of T Cells and Macrophages in Asthma Pathogenesis: A New Perspective on Mutual Crosstalk. Mediators Inflamm 2020; 2020:7835284. [PMID: 32922208 PMCID: PMC7453253 DOI: 10.1155/2020/7835284] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
Asthma is associated with innate and adaptive immunity mediated by immune cells. T cell or macrophage dysfunction plays a particularly significant role in asthma pathogenesis. Furthermore, crosstalk between them continuously transmits proinflammatory or anti-inflammatory signals, causing the immune cell activation or repression in the immune response. Consequently, the imbalanced immune microenvironment is the major cause of the exacerbation of asthma. Here, we discuss the role of T cells, macrophages, and their interactions in asthma pathogenesis.
Collapse
|
50
|
Branchett WJ, O'Garra A, Lloyd CM. Transcriptomic analysis reveals diverse gene expression changes in airway macrophages during experimental allergic airway disease. Wellcome Open Res 2020; 5:101. [PMID: 32587903 PMCID: PMC7309452 DOI: 10.12688/wellcomeopenres.15875.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2020] [Indexed: 12/26/2022] Open
Abstract
Background: Airway macrophages (AMs) are the most abundant leukocytes in the healthy airway lumen and have a highly specialised but plastic phenotype that is governed by signals in the local microenvironment. AMs are thought to maintain immunological homeostasis in the steady state, but have also been implicated in the pathogenesis of allergic airway disease (AAD). In this study, we aimed to better understand these potentially contrasting AM functions using transcriptomic analysis. Methods: Bulk RNA sequencing was performed on AMs (CD11c + Siglec F + CD64 + CD45 + SSC hi) flow cytometry sorted from C57BL/6 mice during experimental AAD driven by repeated house dust mite inhalation (AMs HDM), compared to control AMs from non-allergic mice. Differentially expressed genes were further analysed by hierarchical clustering and biological pathway analysis. Results: AMs HDM showed increased expression of genes associated with antigen presentation, inflammatory cell recruitment and tissue repair, including several chemokine and matrix metalloproteinase genes. This was accompanied by increased expression of mitochondrial electron transport chain subunit genes and the retinoic acid biosynthetic enzyme gene Raldh2. Conversely, AMs HDM displayed decreased expression of a number of cell cycle genes, genes related to cytoskeletal functions and a subset of genes implicated in antimicrobial innate immunity, such as Tlr5, Il18 and Tnf. Differential gene expression in AMs HDM was consistent with upstream effects of the cytokines IL-4 and IFN-γ, both of which were present at increased concentrations in lung tissue after HDM treatment. Conclusions: These data highlight diverse gene expression changes in the total AM population in a clinically relevant mouse model of AAD, collectively suggestive of contributions to inflammation and tissue repair/remodelling, but with decreases in certain steady state cellular and immunological functions.
Collapse
Affiliation(s)
- William J. Branchett
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, W2 1NY, UK
| | - Anne O'Garra
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London, NW1 1AT, UK
| | - Clare M. Lloyd
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, W2 1NY, UK
| |
Collapse
|