51
|
D'Orazio SEF. Innate and Adaptive Immune Responses during Listeria monocytogenes Infection. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0065-2019. [PMID: 31124430 PMCID: PMC11086964 DOI: 10.1128/microbiolspec.gpp3-0065-2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Indexed: 12/15/2022] Open
Abstract
It could be argued that we understand the immune response to infection with Listeria monocytogenes better than the immunity elicited by any other bacteria. L. monocytogenes are Gram-positive bacteria that are genetically tractable and easy to cultivate in vitro, and the mouse model of intravenous (i.v.) inoculation is highly reproducible. For these reasons, immunologists frequently use the mouse model of systemic listeriosis to dissect the mechanisms used by mammalian hosts to recognize and respond to infection. This article provides an overview of what we have learned over the past few decades and is divided into three sections: "Innate Immunity" describes how the host initially detects the presence of L. monocytogenes and characterizes the soluble and cellular responses that occur during the first few days postinfection; "Adaptive Immunity" discusses the exquisitely specific T cell response that mediates complete clearance of infection and immunological memory; "Use of Attenuated Listeria as a Vaccine Vector" highlights the ways that investigators have exploited our extensive knowledge of anti-Listeria immunity to develop cancer therapeutics.
Collapse
Affiliation(s)
- Sarah E F D'Orazio
- University of Kentucky, Microbiology, Immunology & Molecular Genetics, Lexington, KY 40536-0298
| |
Collapse
|
52
|
Teh YC, Ding JL, Ng LG, Chong SZ. Capturing the Fantastic Voyage of Monocytes Through Time and Space. Front Immunol 2019; 10:834. [PMID: 31040854 PMCID: PMC6476989 DOI: 10.3389/fimmu.2019.00834] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 03/29/2019] [Indexed: 02/02/2023] Open
Abstract
Monocytes are a subset of cells that are categorized together with dendritic cells (DCs) and macrophages in the mononuclear phagocyte system (MPS). Despite sharing several phenotypic and functional characteristics with MPS cells, monocytes are unique cells with the ability to function as both precursor and effector cells in their own right. Before the development of hematopoietic stem cells (HSCs) in utero, monocytes are derived from erythro-myeloid precursors (EMPs) in the fetal liver that are important for populating the majority of tissue resident macrophages. After birth, monocytes arise from bone marrow (BM)-derived HSCs and are released into the circulation upon their maturation, where they survey peripheral tissues and maintain endothelial integrity. Upon sensing of microbial breaches or inflammatory stimuli, monocytes migrate into tissues where their plasticity allows them to differentiate into cells that resemble macrophages or DCs according to the environmental niche. Alternatively, they may also migrate into tissues in the absence of inflammation and remain in an undifferentiated state where they perform homeostatic roles. As monocytes are typically on the move, the availability of intravital imaging approaches has provided further insights into their trafficking patterns in distinct tissue compartments. In this review, we outline the importance of understanding their functional behavior in the context of tissue compartments, and how these studies may contribute towards improved vaccine and future therapeutic strategies.
Collapse
Affiliation(s)
- Ye Chean Teh
- Functional Immune Imaging, Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Biopolis, Singapore.,Department of Biological Sciences, National University of Singapore (NUS), Singapore, Singapore
| | - Jeak Ling Ding
- Department of Biological Sciences, National University of Singapore (NUS), Singapore, Singapore
| | - Lai Guan Ng
- Functional Immune Imaging, Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Biopolis, Singapore.,Department of Microbiology & Immunology, Immunology Programme, Life Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Shu Zhen Chong
- Functional Immune Imaging, Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Biopolis, Singapore
| |
Collapse
|
53
|
Eldredge LC, Creasy RS, Presnell S, Debley JS, Juul SE, Mayock DE, Ziegler SF. Infants with evolving bronchopulmonary dysplasia demonstrate monocyte-specific expression of IL-1 in tracheal aspirates. Am J Physiol Lung Cell Mol Physiol 2019; 317:L49-L56. [PMID: 30969811 DOI: 10.1152/ajplung.00060.2019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) remains a devastating consequence of prematurity. Repeated inflammatory insults worsen lung injury, but there are no predictors for BPD-related respiratory outcomes or targeted therapies. We sought to understand inflammatory mechanisms in evolving BPD through molecular characterization of monocytes in tracheal aspirates from infants at risk for developing BPD. We performed flow cytometry targeting myeloid cell populations on prospectively collected tracheal aspirates from intubated patients born before 29 wk of gestation and <30 days old. We identified CD14+CD16+ (double-positive) and CD14+CD16- (single-positive) monocytes and characterized their gene expression profiles by RNA sequencing and quantitative PCR. We further analyzed differential gene expression between time points to evaluate changes in monocyte function over the first weeks of life. Expression of IL-1A, IL-1B, and IL-1 receptor antagonist mRNA was increased in monocytes collected at day of life (DOL) 7, DOL 14, and DOL 28 compared with those collected at DOL 3. This study suggests that early changes in monocyte-specific IL-1 cytokine pathways may be associated with evolving BPD.
Collapse
Affiliation(s)
- Laurie C Eldredge
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington
| | - Rane S Creasy
- Immunology Program, Benaroya Research Institute , Seattle, Washington
| | - Scott Presnell
- Bioinformatics Program, Benaroya Research Institute , Seattle, Washington
| | - Jason S Debley
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington.,Center for Immunity and Immunotherapies, Seattle Children's Research Institute , Seattle, Washington
| | - Sandra E Juul
- Division of Neonatology, Department of Pediatrics, University of Washington , Seattle, Washington
| | - Dennis E Mayock
- Division of Neonatology, Department of Pediatrics, University of Washington , Seattle, Washington
| | - Steven F Ziegler
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, Washington
| |
Collapse
|
54
|
Ben-David-Naim M, Dagan A, Grad E, Aizik G, Nordling-David MM, Morss Clyne A, Granot Z, Golomb G. Targeted siRNA Nanoparticles for Mammary Carcinoma Therapy. Cancers (Basel) 2019; 11:E442. [PMID: 30934857 PMCID: PMC6521050 DOI: 10.3390/cancers11040442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
Non-viral, polymeric-based, siRNA nanoparticles (NPs) have been proposed as promising gene delivery systems. Encapsulating siRNA in targeted NPs could confer improved biological stability, extended half-life, enhanced permeability, effective tumor accumulation, and therapy. In this work, a peptide derived from apolipoprotein B100 (ApoB-P), the protein moiety of low-density lipoprotein, was used to target siRNA-loaded PEGylated NPs to the extracellular matrix/proteoglycans (ECM/PGs) of a mammary carcinoma tumor. siRNA against osteopontin (siOPN), a protein involved in breast cancer development and progression, was encapsulated into PEGylated poly(d,l-lactic-co-glycolic acid) (PLGA) NPs using the double emulsion solvent diffusion technique. The NPs obtained possessed desired physicochemical properties including ~200 nm size, a neutral surface charge, and high siOPN loading of ~5 µg/mg. ApoB-P-targeted NPs exhibited both enhanced binding to isolated ECM and internalization by MDA-MB-231 human mammary carcinoma cells, in comparison to non-targeted NPs. Increased accumulation of the targeted NPs was achieved in the primary mammary tumor of mice xenografted with MDA-MB-231 mammary carcinoma cells as well as in the lungs, one of the main sites affected by metastases. siOPN NPs treatment resulted in significant inhibition of tumor growth (similar bioactivity of both formulations), accompanied with significant reduction of OPN mRNA levels (~40% knockdown of mRNA levels). We demonstrated that targeted NPs possessed enhanced tumor accumulation with increased therapeutic potential in mice models of mammary carcinoma.
Collapse
Affiliation(s)
- Meital Ben-David-Naim
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Arie Dagan
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Etty Grad
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Gil Aizik
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Mirjam M Nordling-David
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Alisa Morss Clyne
- Department of Mechanical Engineering and Mechanics, Drexel University, Philadelphia, PA 19104, USA.
| | - Zvi Granot
- Institute for Medical Research Israel Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| | - Gershon Golomb
- Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel.
| |
Collapse
|
55
|
Guilbaud E, Gautier EL, Yvan-Charvet L. Macrophage Origin, Metabolic Reprogramming and IL-1 Signaling: Promises and Pitfalls in Lung Cancer. Cancers (Basel) 2019; 11:E298. [PMID: 30832375 PMCID: PMC6468621 DOI: 10.3390/cancers11030298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/21/2019] [Accepted: 02/26/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophages are tissue-resident cells that act as immune sentinels to maintain tissue integrity, preserve self-tolerance and protect against invading pathogens. Lung macrophages within the distal airways face around 8000⁻9000 L of air every day and for that reason are continuously exposed to a variety of inhaled particles, allergens or airborne microbes. Chronic exposure to irritant particles can prime macrophages to mediate a smoldering inflammatory response creating a mutagenic environment and favoring cancer initiation. Tumor-associated macrophages (TAMs) represent the majority of the tumor stroma and maintain intricate interactions with malignant cells within the tumor microenvironment (TME) largely influencing the outcome of cancer growth and metastasis. A number of macrophage-centered approaches have been investigated as potential cancer therapy and include strategies to limit their infiltration or exploit their antitumor effector functions. Recently, strategies aimed at targeting IL-1 signaling pathway using a blocking antibody have unexpectedly shown great promise on incident lung cancer. Here, we review the current understanding of the bridge between TAM metabolism, IL-1 signaling, and effector functions in lung adenocarcinoma and address the challenges to successfully incorporating these pathways into current anticancer regimens.
Collapse
Affiliation(s)
- Emma Guilbaud
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France.
| | - Emmanuel L Gautier
- Institut National de la Santé et de la Recherche Médicale (Inserm) UMR_S 1166, Sorbonnes Universités, Hôpital de la Pitié Salpêtrière, 75013 Paris, France.
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, 06204 Nice, France.
| |
Collapse
|
56
|
Looney MR, Headley MB. Live imaging of the pulmonary immune environment. Cell Immunol 2018; 350:103862. [PMID: 30336937 DOI: 10.1016/j.cellimm.2018.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 07/12/2018] [Accepted: 09/27/2018] [Indexed: 01/22/2023]
Abstract
The lung represents a unique immune environment. The primary function of the lung is to enable gas exchange by facilitating the transfer of oxygen into and carbon dioxide out of the blood. However, as a direct byproduct of this process the lung is also constantly exposed to particles, allergens, and pathogens alongside air itself. Due to this, the pulmonary immune system exists in a fine balance between quiescence and inflammation, deviations from which can lead to a failure in respiratory function. A rich history exists attempting to define the critical features of lung immunity, and most recently advances in intravital microscopy have enabled the visualization of intercellular immune dynamics in both steady-state and a variety of disease conditions. In this review, we will summarize a variety of approaches to intravital lung imaging as well as how its application has advanced our understanding of normal lung function as well as disease states such as pulmonary metastasis, asthma, and lung injury.
Collapse
Affiliation(s)
- Mark R Looney
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA; Department of Laboratory Medicine, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Mark B Headley
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| |
Collapse
|
57
|
Loyher PL, Hamon P, Laviron M, Meghraoui-Kheddar A, Goncalves E, Deng Z, Torstensson S, Bercovici N, Baudesson de Chanville C, Combadière B, Geissmann F, Savina A, Combadière C, Boissonnas A. Macrophages of distinct origins contribute to tumor development in the lung. J Exp Med 2018; 215:2536-2553. [PMID: 30201786 PMCID: PMC6170177 DOI: 10.1084/jem.20180534] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 07/02/2018] [Accepted: 08/28/2018] [Indexed: 01/22/2023] Open
Abstract
Tissue-resident macrophages can self-maintain without contribution of adult hematopoiesis. Herein we show that tissue-resident interstitial macrophages (Res-TAMs) in mouse lungs contribute to the pool of tumor-associated macrophages (TAMs) together with CCR2-dependent recruited macrophages (MoD-TAMs). Res-TAMs largely correlated with tumor cell growth in vivo, while MoD-TAMs accumulation was associated with enhanced tumor spreading. Both cell subsets were depleted after chemotherapy, but MoD-TAMs rapidly recovered and performed phagocytosis-mediated tumor clearance. Interestingly, anti-VEGF treatment combined with chemotherapy inhibited both Res and Mod-TAM reconstitution without affecting monocyte infiltration and improved its efficacy. Our results reveal that the developmental origin of TAMs dictates their relative distribution, function, and response to cancer therapies in lung tumors.
Collapse
Affiliation(s)
- Pierre-Louis Loyher
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France.,Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pauline Hamon
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Marie Laviron
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Aïda Meghraoui-Kheddar
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Elena Goncalves
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Zihou Deng
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sara Torstensson
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Nadège Bercovici
- Inserm, U1016, Institut Cochin, CNRS UMR8104, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Camille Baudesson de Chanville
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Béhazine Combadière
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Frederic Geissmann
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ariel Savina
- Institut Roche, 30, Boulogne-Billancourt Cedex, France
| | - Christophe Combadière
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Alexandre Boissonnas
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| |
Collapse
|
58
|
Adam L, López-González M, Björk A, Pålsson S, Poux C, Wahren-Herlenius M, Fernández C, Spetz AL. Early Resistance of Non-virulent Mycobacterial Infection in C57BL/6 Mice Is Associated With Rapid Up-Regulation of Antimicrobial Cathelicidin Camp. Front Immunol 2018; 9:1939. [PMID: 30233570 PMCID: PMC6129578 DOI: 10.3389/fimmu.2018.01939] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/06/2018] [Indexed: 01/04/2023] Open
Abstract
Early clearance of tuberculosis is the successful eradication of inhaled bacteria before the development of an adaptive immune response. We previously showed, by utilizing a non-virulent mycobacteria infection model, that C57BL/6 mice are more efficient than BALB/c in their control of bacterial growth in the lungs during the first weeks of the infection. Here, we assessed early (within 1–3 days) innate immune events locally in the lungs to identify factors that may contribute to the control of non-virulent mycobacterial burden. We confirmed that C57BL/6 mice are more resistant to infection compared with BALB/c after intranasal inoculation with mycobacterium. Transcriptomic analyses revealed a remarkably silent signature in C57BL/6 mice despite effective control of bacterial growth. In contrast, BALB/c mice up-regulated genes associated with neutrophil and myeloid cell chemotaxis and migration. Flow cytometry analyses corroborated the transcriptomic analyses and demonstrated influx of both neutrophil and myeloid cell populations in BALB/c mice, while these did not increase in C57BL/6 mice. We further detected increased release of TNF-α from BALB/c lung cells but limited release from C57BL/6-derived cells. However, C57BL/6 mice showed a marked early up-regulation of the Camp gene, encoding the cathelicidin CRAMP peptide, post-mycobacterial exposure. CRAMP (LL-37 in human) expression in the lungs was confirmed using immunofluorescence staining. Altogether, these findings show that C57BL/6 mice can clear the mycobacterial infection early and that this early control is associated with high CRAMP expression in the lungs without concomitant influx of immune cells. The role of CRAMP/LL-37 during mycobacterial infection may be relevant for novel protective strategies, and warrants further studies of human cohorts.
Collapse
Affiliation(s)
- Lucille Adam
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Moisés López-González
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Albin Björk
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sandra Pålsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Candice Poux
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Marie Wahren-Herlenius
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Carmen Fernández
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Anna-Lena Spetz
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| |
Collapse
|
59
|
Segmented Filamentous Bacteria Provoke Lung Autoimmunity by Inducing Gut-Lung Axis Th17 Cells Expressing Dual TCRs. Cell Host Microbe 2018; 22:697-704.e4. [PMID: 29120746 DOI: 10.1016/j.chom.2017.10.007] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 07/04/2017] [Accepted: 10/16/2017] [Indexed: 01/19/2023]
Abstract
Lung complications are a major cause of rheumatoid arthritis-related mortality. Involvement of gut microbiota in lung diseases by the gut-lung axis has been widely observed, but the underlying mechanism remains mostly unknown. Using an autoimmune arthritis model, we show that a constituent of the gut microbiota, segmented filamentous bacteria (SFB), distantly provoke lung pathology. SFB induce autoantibodies in lung during the pre-arthritic phase, and SFB-dependent lung pathology requires the T helper 17 (Th17) responses. SFB-induced gut Th17 cells are preferentially recruited to lung over spleen due to robust expression in the lung of the Th17 chemoattractant, CCL20. Additionally, we found that in peripheral tissues, SFB selectively expand dual T cell receptor (TCR)-expressing Th17 cells recognizing both an SFB epitope and self-antigen, thus augmenting autoimmunity. This study reveals mechanisms for commensal-mediated gut-lung crosstalk and dual TCR-based autoimmunity.
Collapse
|
60
|
Joshi N, Walter JM, Misharin AV. Alveolar Macrophages. Cell Immunol 2018; 330:86-90. [DOI: 10.1016/j.cellimm.2018.01.005] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/07/2018] [Accepted: 01/11/2018] [Indexed: 12/15/2022]
|
61
|
Catacchio I, Scattone A, Silvestris N, Mangia A. Immune Prophets of Lung Cancer: The Prognostic and Predictive Landscape of Cellular and Molecular Immune Markers. Transl Oncol 2018; 11:825-835. [PMID: 29729581 PMCID: PMC6050352 DOI: 10.1016/j.tranon.2018.04.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths throughout the world. The majority of patients are diagnosed with locally advanced or metastatic disease when surgery, the best curative option, is no longer feasible. Thus, the prognosis of lung cancer remains poor and heterogeneous and new biomarkers are needed. As the immune system plays a pivotal role in cancer, the study of tumor microenvironment, with regard to the immune component, may provide valuable information for a better comprehension of the pathogenesis and progression of the disease. Through a detailed and critical evaluation of the most recent publications on this topic, we provide evidences of the prognostic and predictive significance of immune markers in tumor and in peripheral blood of lung cancer patients: from the landscape of immune cells (macrophages, neutrophils, lymphocytes and natural killer) and their cytokines, to the analysis of immune-checkpoints (PD-L1 and CTLA4), up to the genetic and epigenetic regulation of the immune response (immune gene signatures and miRNA). We also argue about the lights and shadows related to immune marker use in clinical practice, emphasizing on one hand the importance of their assessment in the choice of therapeutic treatment, on the other, the difficulty in their determination and reproducibility of literature data. The following review gives a foundation and a suggestion for future studies investigating tumor immunology in lung cancer.
Collapse
Affiliation(s)
- Ivana Catacchio
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori, Bari 70124, Italy
| | - Anna Scattone
- Pathology Department, IRCCS-Istituto Tumori, Bari 70124, Italy
| | | | - Anita Mangia
- Functional Biomorphology Laboratory, IRCCS-Istituto Tumori, Bari 70124, Italy.
| |
Collapse
|
62
|
Kiss M, Van Gassen S, Movahedi K, Saeys Y, Laoui D. Myeloid cell heterogeneity in cancer: not a single cell alike. Cell Immunol 2018; 330:188-201. [PMID: 29482836 DOI: 10.1016/j.cellimm.2018.02.008] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/10/2018] [Accepted: 02/11/2018] [Indexed: 12/14/2022]
Abstract
Tumors of various histological origins show abundant infiltration of myeloid cells from early stages of disease progression. These cells have a profound impact on antitumor immunity and influence fundamental processes that underlie malignancy, including neoangiogenesis, sustained cancer cell proliferation, metastasis and therapy resistance. For these reasons, development of therapeutic approaches to deplete or reprogram myeloid cells in cancer is an emerging field of interest. However, knowledge about the heterogeneity of myeloid cells in tumors and their variability between patients and disease stages is still limited. In this review, we summarize the most recent advances in our understanding about how the phenotype of tumor-associated macrophages, monocytes, neutrophils, myeloid-derived suppressor cells and dendritic cells is dictated by their ontogeny, activation status and localization. We also outline major open questions that will only be resolved by applying high-dimensional single-cell technologies and systems biology approaches in the analysis of the tumor microenvironment.
Collapse
Affiliation(s)
- Mate Kiss
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium.
| | - Sofie Van Gassen
- IDLab, Department of Information Technology, Ghent University - IMEC, Ghent, Belgium; Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium
| | - Kiavash Movahedi
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium; Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Damya Laoui
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium; Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium.
| |
Collapse
|
63
|
Hawley CA, Rojo R, Raper A, Sauter KA, Lisowski ZM, Grabert K, Bain CC, Davis GM, Louwe PA, Ostrowski MC, Hume DA, Pridans C, Jenkins SJ. Csf1r-mApple Transgene Expression and Ligand Binding In Vivo Reveal Dynamics of CSF1R Expression within the Mononuclear Phagocyte System. THE JOURNAL OF IMMUNOLOGY 2018; 200:2209-2223. [PMID: 29440354 PMCID: PMC5834790 DOI: 10.4049/jimmunol.1701488] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/17/2018] [Indexed: 01/18/2023]
Abstract
CSF1 is the primary growth factor controlling macrophage numbers, but whether expression of the CSF1 receptor differs between discrete populations of mononuclear phagocytes remains unclear. We have generated a Csf1r-mApple transgenic fluorescent reporter mouse that, in combination with lineage tracing, Alexa Fluor 647–labeled CSF1-Fc and CSF1, and a modified ΔCsf1–enhanced cyan fluorescent protein (ECFP) transgene that lacks a 150 bp segment of the distal promoter, we have used to dissect the differentiation and CSF1 responsiveness of mononuclear phagocyte populations in situ. Consistent with previous Csf1r-driven reporter lines, Csf1r-mApple was expressed in blood monocytes and at higher levels in tissue macrophages, and was readily detectable in whole mounts or with multiphoton microscopy. In the liver and peritoneal cavity, uptake of labeled CSF1 largely reflected transgene expression, with greater receptor activity in mature macrophages than monocytes and tissue-specific expression in conventional dendritic cells. However, CSF1 uptake also differed between subsets of monocytes and discrete populations of tissue macrophages, which in macrophages correlated with their level of dependence on CSF1 receptor signaling for survival rather than degree of transgene expression. A double ΔCsf1r-ECFP-Csf1r-mApple transgenic mouse distinguished subpopulations of microglia in the brain, and permitted imaging of interstitial macrophages distinct from alveolar macrophages, and pulmonary monocytes and conventional dendritic cells. The Csf1r-mApple mice and fluorescently labeled CSF1 will be valuable resources for the study of macrophage and CSF1 biology, which are compatible with existing EGFP-based reporter lines.
Collapse
Affiliation(s)
- Catherine A Hawley
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Rocio Rojo
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Anna Raper
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Kristin A Sauter
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Zofia M Lisowski
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Kathleen Grabert
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Calum C Bain
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Gemma M Davis
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom.,Faculty of Life Sciences, The University of Manchester, Manchester M13 9PL, United Kingdom
| | - Pieter A Louwe
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom
| | - Michael C Ostrowski
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425; and
| | - David A Hume
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom.,The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom.,Mater Research-University of Queensland, Translational Research Institute, Woolloongabba, Queensland 4104, Australia
| | - Clare Pridans
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom.,The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Stephen J Jenkins
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, United Kingdom;
| |
Collapse
|
64
|
Angiogenic factor-driven inflammation promotes extravasation of human proangiogenic monocytes to tumours. Nat Commun 2018; 9:355. [PMID: 29367702 PMCID: PMC5783934 DOI: 10.1038/s41467-017-02610-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/13/2017] [Indexed: 12/20/2022] Open
Abstract
Recruitment of circulating monocytes is critical for tumour angiogenesis. However, how human monocyte subpopulations extravasate to tumours is unclear. Here we show mechanisms of extravasation of human CD14dimCD16+ patrolling and CD14+CD16+ intermediate proangiogenic monocytes (HPMo), using human tumour xenograft models and live imaging of transmigration. IFNγ promotes an increase of the chemokine CX3CL1 on vessel lumen, imposing continuous crawling to HPMo and making these monocytes insensitive to chemokines required for their extravasation. Expression of the angiogenic factor VEGF and the inflammatory cytokine TNF by tumour cells enables HPMo extravasation by inducing GATA3-mediated repression of CX3CL1 expression. Recruited HPMo boosts angiogenesis by secreting MMP9 leading to release of matrix-bound VEGF-A, which amplifies the entry of more HPMo into tumours. Uncovering the extravasation cascade of HPMo sets the stage for future tumour therapies. Circulating myeloid cells can leave the vasculature to infiltrate tumours and are thought to contribute to tumour angiogenesis. Here the authors live image monocytes that migrate to xenograft tumours and map an extravasation cascade of human proangiogenic monocytes into the tumour.
Collapse
|
65
|
Chong SZ, Evrard M, Goh CC, Ng LG. Illuminating the covert mission of mononuclear phagocytes in their regional niches. Curr Opin Immunol 2017; 50:94-101. [PMID: 29275187 DOI: 10.1016/j.coi.2017.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/17/2017] [Accepted: 12/01/2017] [Indexed: 12/22/2022]
Abstract
Monocytes, dendritic cells (DCs) and macrophages have been classically categorized into the mononuclear phagocyte system (MPS) based on their similar functional and phenotypic characteristics. While an increasing amount of research has revealed substantial ontogenic and functional differences among these cells, the reasons behind their heterogeneity and strategic positioning in specific niches throughout the body are yet to be fully elucidated. In this review, we outline how recent advances in intravital imaging studies have dissected this phenomenon and have allowed us to appreciate how MPS cells exploit their regional niches to specialize and maximize their functional properties. Understanding their cellular behavior in each of their specialized microenvironment will eventually allow us to target specific cells and their behavioral patterns for improved vaccine and therapeutic purposes.
Collapse
Affiliation(s)
- Shu Zhen Chong
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore, Singapore.
| | - Maximilien Evrard
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore, Singapore
| | - Chi Ching Goh
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore, Singapore
| | - Lai Guan Ng
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, 138648 Singapore, Singapore.
| |
Collapse
|
66
|
Fröhlich E. Toxicity of orally inhaled drug formulations at the alveolar barrier: parameters for initial biological screening. Drug Deliv 2017; 24:891-905. [PMID: 28574335 PMCID: PMC8241192 DOI: 10.1080/10717544.2017.1333172] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oral delivery is the most common mode of systemic drug application. Inhalation is mainly used for local therapy of lung diseases but may also be a promising route for systemic delivery of drugs that have poor oral bioavailability. The thin alveolar barrier enables fast and efficient uptake of many molecules and could deliver small molecules and proteins, which are susceptible to degradation and show poor absorption by oral application. The low rate of biotransformation and proteolytic degradation increases bioavailability of drugs but accumulation of not absorbed material may impair normal lung function. This limitation is more relevant for compounds that should be systematically active because higher doses have to be applied to the lung. The review describes processes that determine absorption of orally inhaled formulations, namely dissolution in the lung lining fluid and uptake and degradation by alveolar epithelial cells and macrophages. Dissolution testing in simulated lung fluid, screening for cytotoxicity and pro-inflammatory action in respiratory cells and study of macrophage morphology, and phagocytosis can help to identify adverse effects of pulmonary formulations.
Collapse
Affiliation(s)
- Eleonore Fröhlich
- a Center for Medical Research, Medical University of Graz , Graz , Austria.,b Research Center Pharmaceutical Engineering GmbH , Graz , Austria
| |
Collapse
|
67
|
Bohmwald K, Espinoza JA, Pulgar RA, Jara EL, Kalergis AM. Functional Impairment of Mononuclear Phagocyte System by the Human Respiratory Syncytial Virus. Front Immunol 2017; 8:1643. [PMID: 29230219 PMCID: PMC5712212 DOI: 10.3389/fimmu.2017.01643] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/10/2017] [Indexed: 12/18/2022] Open
Abstract
The mononuclear phagocyte system (MPS) comprises of monocytes, macrophages (MΦ), and dendritic cells (DCs). MPS is part of the first line of immune defense against a wide range of pathogens, including viruses, such as the human respiratory syncytial virus (hRSV). The hRSV is an enveloped virus that belongs to the Pneumoviridae family, Orthopneumovirus genus. This virus is the main etiological agent causing severe acute lower respiratory tract infection, especially in infants, children and the elderly. Human RSV can cause bronchiolitis and pneumonia and it has also been implicated in the development of recurrent wheezing and asthma. Monocytes, MΦ, and DCs significantly contribute to acute inflammation during hRSV-induced bronchiolitis and asthma exacerbation. Furthermore, these cells seem to be an important component for the association between hRSV and reactive airway disease. After hRSV infection, the first cells encountered by the virus are respiratory epithelial cells, alveolar macrophages (AMs), DCs, and monocytes in the airways. Because AMs constitute the predominant cell population at the alveolar space in healthy subjects, these cells work as major innate sentinels for the recognition of pathogens. Although adaptive immunity is crucial for viral clearance, AMs are required for the early immune response against hRSV, promoting viral clearance and controlling immunopathology. Furthermore, exposure to hRSV may affect the phagocytic and microbicidal capacity of monocytes and MΦs against other infectious agents. Finally, different studies have addressed the roles of different DC subsets during infection by hRSV. In this review article, we discuss the role of the lung MPS during hRSV infection and their involvement in the development of bronchiolitis.
Collapse
Affiliation(s)
- Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Janyra A Espinoza
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Raúl A Pulgar
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Evelyn L Jara
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
68
|
Plebanek MP, Angeloni NL, Vinokour E, Li J, Henkin A, Martinez-Marin D, Filleur S, Bhowmick R, Henkin J, Miller SD, Ifergan I, Lee Y, Osman I, Thaxton CS, Volpert OV. Pre-metastatic cancer exosomes induce immune surveillance by patrolling monocytes at the metastatic niche. Nat Commun 2017; 8:1319. [PMID: 29105655 PMCID: PMC5673063 DOI: 10.1038/s41467-017-01433-3] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 09/18/2017] [Indexed: 12/11/2022] Open
Abstract
Metastatic cancers produce exosomes that condition pre-metastatic niches in remote microenvironments to favor metastasis. In contrast, here we show that exosomes from poorly metastatic melanoma cells can potently inhibit metastasis to the lung. These "non-metastatic" exosomes stimulate an innate immune response through the expansion of Ly6Clow patrolling monocytes (PMo) in the bone marrow, which then cause cancer cell clearance at the pre-metastatic niche, via the recruitment of NK cells and TRAIL-dependent killing of melanoma cells by macrophages. These events require the induction of the Nr4a1 transcription factor and are dependent on pigment epithelium-derived factor (PEDF) on the outer surface of exosomes. Importantly, exosomes isolated from patients with non-metastatic primary melanomas have a similar ability to suppress lung metastasis. This study thus demonstrates that pre-metastatic tumors produce exosomes, which elicit a broad range of PMo-reliant innate immune responses via trigger(s) of immune surveillance, causing cancer cell clearance at the pre-metastatic niche.
Collapse
Affiliation(s)
- Michael P Plebanek
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
- Simpson-Querrey Institute for Bionantechnology in Medicine, 303 E. Superior St, Chicago, IL, 60611, USA
| | - Nicholas L Angeloni
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
- Simpson-Querrey Institute for Bionantechnology in Medicine, 303 E. Superior St, Chicago, IL, 60611, USA
| | - Elena Vinokour
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Jia Li
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Anna Henkin
- The Department for Health and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, 45 Carleton St., Cambridge, MA, 02139, USA
| | - Dalia Martinez-Marin
- Department of Urology, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX, 79430-6591, USA
| | - Stephanie Filleur
- Department of Urology, Texas Tech University Health Sciences Center, 3601 4th St, Lubbock, TX, 79430-6591, USA
| | - Reshma Bhowmick
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA
| | - Jack Henkin
- Chemistry of Life Processes Institute, Northwestern University, 2145 Sheridan Rd, Evanston, IL, 60208, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Igal Ifergan
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Yesung Lee
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, 240 East 38th Street, New York, NY, 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University, Langone Medical Center, 160 East 34th Street, New York, NY, 10016, USA
| | - Iman Osman
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, 240 East 38th Street, New York, NY, 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University, Langone Medical Center, 160 East 34th Street, New York, NY, 10016, USA
| | - C Shad Thaxton
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
- Simpson-Querrey Institute for Bionantechnology in Medicine, 303 E. Superior St, Chicago, IL, 60611, USA
- Northwestern University International Institute for Nanotechnology, 2145 Sheridan Rd., Evanston, IL, 60208, USA
| | - Olga V Volpert
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, 1881 East Rd, Houston, TX, 77054, USA.
| |
Collapse
|
69
|
Alternative splicing of hepatitis B virus: A novel virus/host interaction altering liver immunity. J Hepatol 2017; 67:687-699. [PMID: 28600137 PMCID: PMC6433284 DOI: 10.1016/j.jhep.2017.05.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 05/24/2017] [Accepted: 05/30/2017] [Indexed: 12/25/2022]
Abstract
BACKGROUND & AIMS Hepatitis B virus (HBV) RNA can undergo alternative splicing, but the relevance of this post-transcriptional regulation remains elusive. The mechanism of HBV alternative splicing regulation and its impact on liver pathogenesis were investigated. METHODS HBV RNA-interacting proteins were identified by RNA pull-down, combined with mass spectrometry analysis. HBV splicing regulation was investigated in chemically and surgically induced liver damage, in whole HBV genome transgenic mice and in hepatoma cells. Viral and endogenous gene expression were quantified by quantitative reverse transcription polymerase chain reaction, Western blot and enzyme-linked immunosorbent assay. Resident liver immune cells were studied by fluorescence-activated cell sorting. RESULTS HBV pregenomic RNA-interacting proteins were identified and 15% were directly related to the splicing machinery. Expression of these splicing factors was modulated in HBV transgenic mice with liver injuries and contributed to an increase of the HBV spliced RNA encoding for HBV splicing-generated protein (HBSP). HBSP transgenic mice with chemically induced liver fibrosis exhibited attenuated hepatic damage. The protective effect of HBSP resulted from a decrease of inflammatory monocyte/macrophage recruitment through downregulation of C-C motif chemokine ligand 2 (CCL2) expression in hepatocytes. In human hepatoma cells, the ability of HBSP to control CCL2 expression was confirmed and maintained in a whole HBV context. Finally, viral spliced RNA detection related to a decrease of CCL2 expression in the livers of HBV chronic carriers underscored this mechanism. CONCLUSION The microenvironment, modified by liver injury, increased HBSP RNA expression through splicing factor regulation, which in turn controlled hepatocyte chemokine synthesis. This feedback mechanism provides a novel insight into liver immunopathogenesis during HBV infection. Lay summary: Hepatitis B virus persists for decades in the liver of chronically infected patients. Immune escape is one of the main mechanisms developed by this virus to survive. Our study highlights how the crosstalk between virus and liver infected cells may contribute to this immune escape.
Collapse
|
70
|
Vroman H, Hendriks RW, Kool M. Dendritic Cell Subsets in Asthma: Impaired Tolerance or Exaggerated Inflammation? Front Immunol 2017; 8:941. [PMID: 28848549 PMCID: PMC5552666 DOI: 10.3389/fimmu.2017.00941] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
Asthma is a prevalent chronic heterogeneous inflammatory disease of the airways, leading to reversible airway obstruction, in which various inflammatory responses can be observed. Mild to moderate asthma patients often present with a Th2-mediated eosinophilic inflammation whereas in severe asthma patients, a Th17-associated neutrophilic or combined Th2 and Th17-mediated eosinophilic/neutrophilic inflammation is observed. The differentiation of these effector Th2 and Th17-cells is induced by allergen-exposed dendritic cells (DCs) that migrate toward the lung draining lymph node. The DC lineage comprises conventional DCs (cDCs) and plasmacytoid DCs (pDCs), of which the cDC lineage consists of type 1 cDCs (cDC1s) and cDC2s. During inflammation, also monocytes can differentiate into so-called monocyte-derived DCs (moDCs). These DC subsets differ both in ontogeny, localization, and in their functional properties. New identification tools and the availability of transgenic mice targeting specific DC subsets enable the investigation of how these different DC subsets contribute to or suppress asthma pathogenesis. In this review, we will discuss mechanisms used by different DC subsets to elicit or hamper the pathogenesis of both Th2-mediated eosinophilic asthma and more severe Th17-mediated neutrophilic inflammation.
Collapse
Affiliation(s)
- Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Mirjam Kool
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
71
|
Amsellem V, Abid S, Poupel L, Parpaleix A, Rodero M, Gary-Bobo G, Latiri M, Dubois-Rande JL, Lipskaia L, Combadiere C, Adnot S. Roles for the CX3CL1/CX3CR1 and CCL2/CCR2 Chemokine Systems in Hypoxic Pulmonary Hypertension. Am J Respir Cell Mol Biol 2017; 56:597-608. [PMID: 28125278 DOI: 10.1165/rcmb.2016-0201oc] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Monocytes/macrophages are major effectors of lung inflammation associated with various forms of pulmonary hypertension (PH). Interactions between the CCL2/CCR2 and CX3CL1/CX3CR1 chemokine systems that guide phagocyte infiltration are incompletely understood. Our objective was to explore the individual and combined actions of CCL2/CCR2 and CX3CL1/CX3CR1 in hypoxia-induced PH in mice; particularly their roles in monocyte trafficking, macrophage polarization, and pulmonary vascular remodeling. The development of hypoxia-induced PH was associated with marked increases in lung levels of CX3CR1, CCR2, and their respective ligands, CX3CL1 and CCL2. Flow cytometry revealed that both inflammatory Ly6Chi and resident Ly6Clo monocyte subsets exhibited sustained increases in blood and a transient peak in lung tissue, and that lung perivascular and alveolar macrophage counts showed sustained elevations. CX3CR1-/- mice were protected against hypoxic PH compared with wild-type mice, whereas CCL2-/- mice and double CX3CR1-/-/CCL2-/- mice exhibited similar PH severity, as did wild-type mice. The protective effects of CX3CR1 deficiency occurred concomitantly with increases in lung monocyte and macrophage counts and with a change from M2 to M1 macrophage polarization that markedly diminished the ability of conditioned media to induce pulmonary artery smooth muscle cell (PA-SMC) proliferation, which was partly dependent on CX3CL1 secretion. Results in mice given the CX3CR1 inhibitor F1 were similar to those in CX3CR1-/- mice. In conclusion, CX3CR1 deficiency protects against hypoxia-induced PH by modulating monocyte recruitment, macrophage polarization, and PA-SMC cell proliferation. Targeting CX3CR1 may hold promise for treating PH.
Collapse
Affiliation(s)
- Valérie Amsellem
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Shariq Abid
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Lucie Poupel
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Aurélien Parpaleix
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Mathieu Rodero
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Guillaume Gary-Bobo
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Mehdi Latiri
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Jean-Luc Dubois-Rande
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Larissa Lipskaia
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Christophe Combadiere
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| | - Serge Adnot
- INSERM U955 and Département de Physiologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Départements Hospitalo Universitaires Ageing Thorax-Vessels-Blood, 94010, Créteil, France; Université Paris-Est Créteil, France; and Sorbonne Universités, Université Pierre et Marie Curie-Université Paris 06, Inserm, UMRS1135, CNRS, Equipes de Recherche Labellisées 8255, Centre d'Immunologie et des Maladies Infectieuses, Paris, France
| |
Collapse
|
72
|
Transcriptional mechanisms that control expression of the macrophage colony-stimulating factor receptor locus. Clin Sci (Lond) 2017; 131:2161-2182. [DOI: 10.1042/cs20170238] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/22/2017] [Accepted: 06/11/2017] [Indexed: 12/17/2022]
Abstract
The proliferation, differentiation, and survival of cells of the macrophage lineage depends upon signals from the macrophage colony-stimulating factor (CSF) receptor (CSF1R). CSF1R is expressed by embryonic macrophages and induced early in adult hematopoiesis, upon commitment of multipotent progenitors to the myeloid lineage. Transcriptional activation of CSF1R requires interaction between members of the E26 transformation-specific family of transcription factors (Ets) (notably PU.1), C/EBP, RUNX, AP-1/ATF, interferon regulatory factor (IRF), STAT, KLF, REL, FUS/TLS (fused in sarcoma/ranslocated in liposarcoma) families, and conserved regulatory elements within the mouse and human CSF1R locus. One element, the Fms-intronic regulatory element (FIRE), within intron 2, is conserved functionally across all the amniotes. Lineage commitment in multipotent progenitors also requires down-regulation of specific transcription factors such as MYB, FLI1, basic leucine zipper transcriptional factor ATF-like (BATF3), GATA-1, and PAX5 that contribute to differentiation of alternative lineages and repress CSF1R transcription. Many of these transcription factors regulate each other, interact at the protein level, and are themselves downstream targets of CSF1R signaling. Control of CSF1R transcription involves feed–forward and feedback signaling in which CSF1R is both a target and a participant; and dysregulation of CSF1R expression and/or function is associated with numerous pathological conditions. In this review, we describe the regulatory network behind CSF1R expression during differentiation and development of cells of the mononuclear phagocyte system.
Collapse
|
73
|
Suijkerbuijk SJE, van Rheenen J. From good to bad: Intravital imaging of the hijack of physiological processes by cancer cells. Dev Biol 2017; 428:328-337. [PMID: 28473106 DOI: 10.1016/j.ydbio.2017.04.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 12/23/2022]
Abstract
Homeostasis of tissues is tightly regulated at the cellular, tissue and organismal level. Interestingly, tumor cells have found ways to hijack many of these physiological processes at all the different levels. Here we review how intravital microscopy techniques have provided new insights into our understanding of tissue homeostasis and cancer progression. In addition, we highlight the different strategies that tumor cells have adopted to use these physiological processes for their own benefit. We describe how visualization of these dynamic processes in living mice has broadened to our view on cancer initiation and progression.
Collapse
Affiliation(s)
- Saskia J E Suijkerbuijk
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands
| | - Jacco van Rheenen
- Hubrecht Institute - KNAW (Royal Netherlands Academy of Arts and Sciences) and University Medical Center Utrecht, 3584 CT Utrecht, The Netherlands; Cancer Genomics Netherlands, 3584 CG Utrecht, The Netherlands.
| |
Collapse
|
74
|
Abstract
Multiphoton microscopy has revealed important insights into cellular behavior in vivo. However, its application in infectious settings often encounters technical, safety and regulatory limitations that prevent its wider use with highly virulent human pathogens. Herein, we present a method that renders multiphoton microscopy in vivo compatible with biosafety level 3 regulations and present an example of its application and potential to visualize a Mycobacterium tuberculosis infection of the mouse lung.
Collapse
|
75
|
Jones GS, D'Orazio SEF. Monocytes Are the Predominant Cell Type Associated with Listeria monocytogenes in the Gut, but They Do Not Serve as an Intracellular Growth Niche. THE JOURNAL OF IMMUNOLOGY 2017; 198:2796-2804. [PMID: 28213502 DOI: 10.4049/jimmunol.1602076] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022]
Abstract
After foodborne transmission of the facultative intracellular bacterial pathogen Listeria monocytogenes, most of the bacterial burden in the gut is extracellular. However, we previously demonstrated that intracellular replication in an as yet unidentified cell type was essential for dissemination and systemic spread of L. monocytogenes In this article, we show that the vast majority of cell-associated L. monocytogenes in the gut were adhered to Ly6Chi monocytes, a cell type that inefficiently internalized L. monocytogenes With bone marrow-derived in vitro cultures, high multiplicity of infection or the use of opsonized bacteria enhanced uptake of L. monocytogenes in CD64- monocytes, but very few bacteria reached the cell cytosol. Surprisingly, monocytes that had upregulated CD64 expression in transition toward becoming macrophages fully supported intracellular growth of L. monocytogenes In contrast, inflammatory monocytes that had increased CD64 expression in the bone marrow of BALB/c/By/J mice prior to L. monocytogenes exposure in the gut did not support L. monocytogenes growth. Thus, contrary to the perception that L. monocytogenes can infect virtually all cell types, neither naive nor inflammatory Ly6Chi monocytes served as a productive intracellular growth niche for L. monocytogenes. These results have broad implications for innate immune recognition of L. monocytogenes in the gut and highlight the need for additional studies on the interaction of extracellular, adherent L. monocytogenes with the unique subsets of myeloid-derived inflammatory cells that infiltrate sites of infection.
Collapse
Affiliation(s)
- Grant S Jones
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40536
| | - Sarah E F D'Orazio
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, KY 40536
| |
Collapse
|
76
|
CX3CR1-dependent endothelial margination modulates Ly6C high monocyte systemic deployment upon inflammation in mice. Blood 2016; 129:1296-1307. [PMID: 28011675 DOI: 10.1182/blood-2016-08-732164] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 12/16/2016] [Indexed: 12/24/2022] Open
Abstract
Two subsets of blood monocytes are commonly described in mice and humans: the classical inflammatory monocytes, which are rapidly mobilized upon inflammation in a CC-chemokine receptor 2-dependent manner, and the nonclassical blood resident monocyte subset that patrols the intraluminal side of the endothelium. Old reports suggest that blood monocytes are distributed into circulating and marginating pools, but no direct evidence of the latter has been obtained so far. Using a combination of in vivo real-time imaging and blood/tissue partitioning by intravascular staining of leukocytes, we showed that both inflammatory and resident monocytes are retained in the bone marrow vasculature, representing an important reservoir of marginated monocytes. Upon lipopolysaccharide or cecal ligation and puncture-induced peritonitis, these marginated cells are rapidly released and recruited to the peritoneum membrane lumen vasculature where they reside through CX3C-chemokine receptor 1 (CX3CR1)-dependent adherence. At a later time point, inflammatory monocytes infiltrate the spleen parenchyma but remain mainly intravascular in the vicinity of the lungs and the peritoneum. Our results show that this monocyte deployment is controlled by a CX3CR1-dependent balance between marginating and circulating monocytes and highlight that tissue infiltration is not a mandatory fate for inflammatory monocytes.
Collapse
|
77
|
Sen D, Jones SM, Oswald EM, Pinkard H, Corbin K, Krummel MF. Tracking the Spatial and Functional Gradient of Monocyte-To-Macrophage Differentiation in Inflamed Lung. PLoS One 2016; 11:e0165064. [PMID: 27755611 PMCID: PMC5068774 DOI: 10.1371/journal.pone.0165064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/05/2016] [Indexed: 12/30/2022] Open
Abstract
Myeloid-derived cells such as monocytes, dendritic cells (DCs), and macrophages are at the heart of the immune effector function in an inflammatory response. But because of the lack of an efficient imaging system to trace these cells live during their migration and maturation in their native environment at sub-cellular resolution, our knowledge is limited to data available from specific time-points analyzed by flow cytometry, histology, genomics and other immunological methods. Here, we have developed a ratiometric imaging method for measuring monocyte maturation in inflamed mouse lungs in situ using real-time using 2-photon imaging and complementary methods. We visualized that while undifferentiated monocytes were predominantly found only in the vasculature, a semi-differentiated monocyte/macrophage population could enter the tissue and resembled more mature and differentiated populations by morphology and surface phenotype. As these cells entered and differentiated, they were already selectively localized near inflamed airways and their entry was associated with changes in motility and morphology. We were able to visualize these during the act of differentiation, a process that can be demonstrated in this way to be faster on a per-cell basis under inflammatory conditions. Finally, our in situ analyses demonstrated increases, in the differentiating cells, for both antigen uptake and the ability to mediate interactions with T cells. This work, while largely confirming proposed models for in situ differentiation, provides important in situ data on the coordinated site-specific recruitment and differentiation of these cells and helps elaborate the predominance of immune pathology at the airways. Our novel imaging technology to trace immunogenic cell maturation in situ will complement existing information available on in situ differentiation deduced from other immunological methods, and assist better understanding of the spatio-temporal cellular behavior during an inflammatory response.
Collapse
Affiliation(s)
- Debasish Sen
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Stephen M. Jones
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Erin M. Oswald
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Henry Pinkard
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Kaitlin Corbin
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| | - Matthew F. Krummel
- Department of Pathology, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
78
|
Barron L, Gharib SA, Duffield JS. Lung Pericytes and Resident Fibroblasts: Busy Multitaskers. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2519-31. [PMID: 27555112 DOI: 10.1016/j.ajpath.2016.07.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 06/30/2016] [Accepted: 07/05/2016] [Indexed: 02/06/2023]
Abstract
Pericytes, resident fibroblasts, and mesenchymal stem cells are poorly described cell populations. They have recently been characterized in much greater detail in rodent lungs and have been shown to play important roles in development, homeostasis, response to injury and pathogens, as well as recovery from damage. These closely related mesenchymal cell populations form extensive connections to the lung's internal structure, as well as its internal and external surfaces. They generate and remodel extracellular matrix, coregulate the vasculature, help maintain and restore the epithelium, and act as sentries for the immune system. In this review, we revisit these functions in light of significant advances in characterizing and tracking lung fibroblast populations in rodents. Lineage tracing experiments have mapped the heritage, identified functions that discriminate lung pericytes from resident fibroblasts, identified a subset of mesenchymal stem cells, and shown these populations to be the predominant progenitors of pathological fibroblasts and myofibroblasts in lung diseases. These findings point to the importance of resident lung mesenchymal populations as therapeutic targets in acute lung injury as well as fibrotic and degenerative diseases. Far from being passive and quiescent, pericytes and resident fibroblasts are busily sensing and responding, through diverse mechanisms, to changes in lung health and function.
Collapse
Affiliation(s)
- Luke Barron
- Department of Research and Development, Biogen, Cambridge, Massachusetts
| | - Sina A Gharib
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington
| | - Jeremy S Duffield
- Department of Research and Development, Biogen, Cambridge, Massachusetts; Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, Washington.
| |
Collapse
|
79
|
Pridans C, Davis GM, Sauter KA, Lisowski ZM, Corripio-Miyar Y, Raper A, Lefevre L, Young R, McCulloch ME, Lillico S, Milne E, Whitelaw B, Hume DA. A Csf1r-EGFP Transgene Provides a Novel Marker for Monocyte Subsets in Sheep. THE JOURNAL OF IMMUNOLOGY 2016; 197:2297-305. [PMID: 27521343 PMCID: PMC5009875 DOI: 10.4049/jimmunol.1502336] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 07/15/2016] [Indexed: 12/12/2022]
Abstract
Expression of Csf1r in adults is restricted to cells of the macrophage lineage. Transgenic reporters based upon the Csf1r locus require inclusion of the highly conserved Fms-intronic regulatory element for expression. We have created Csf1r-EGFP transgenic sheep via lentiviral transgenesis of a construct containing elements of the mouse Fms-intronic regulatory element and Csf1r promoter. Committed bone marrow macrophage precursors and blood monocytes express EGFP in these animals. Sheep monocytes were divided into three populations, similar to classical, intermediate, and nonclassical monocytes in humans, based upon CD14 and CD16 expression. All expressed EGFP, with increased levels in the nonclassical subset. Because Csf1r expression coincides with the earliest commitment to the macrophage lineage, Csf1r-EGFP bone marrow provides a tool for studying the earliest events in myelopoiesis using the sheep as a model.
Collapse
Affiliation(s)
- Clare Pridans
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Gemma M Davis
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Kristin A Sauter
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Zofia M Lisowski
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | | | - Anna Raper
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Lucas Lefevre
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Rachel Young
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Mary E McCulloch
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Simon Lillico
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - Elspeth Milne
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian EH25 9RG, United Kingdom
| | - Bruce Whitelaw
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| | - David A Hume
- The Roslin Institute, University of Edinburgh, Midlothian EH25 9RG, United Kingdom; and
| |
Collapse
|
80
|
|
81
|
Nourshargh S, Renshaw SA, Imhof BA. Reverse Migration of Neutrophils: Where, When, How, and Why? Trends Immunol 2016; 37:273-286. [PMID: 27055913 DOI: 10.1016/j.it.2016.03.006] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/14/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023]
Abstract
Neutrophil migration to injured and pathogen-infected tissues is a fundamental component of innate immunity. An array of cellular and molecular events mediate this response to collectively guide neutrophils out of the vasculature and towards the core of the ensuing inflammatory reaction where they exert effector functions. Advances in imaging modalities have revealed that neutrophils can also exhibit motility away from sites of inflammation and injury, although it is unclear under what circumstances this reverse migration is a physiological protective response, and when it has pathophysiological relevance. Here we review different types of neutrophil reverse migration and discuss the current understanding of the associated mechanisms. In this context we propose clarifications to the existing terminology used to describe the many facets of neutrophil reverse migration.
Collapse
Affiliation(s)
- Sussan Nourshargh
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Stephen A Renshaw
- Bateson Centre and Department of Infection, Immunity and Cardiovascular Disease, Firth Court, University of Sheffield, Western Bank, Sheffield S10 2TN, UK.
| | - Beat A Imhof
- Centre Médical Universitaire, Rue Michel-Servet 1, Geneva 1211, Switzerland.
| |
Collapse
|
82
|
Hoffmann F, Ender F, Schmudde I, Lewkowich IP, Köhl J, König P, Laumonnier Y. Origin, Localization, and Immunoregulatory Properties of Pulmonary Phagocytes in Allergic Asthma. Front Immunol 2016; 7:107. [PMID: 27047494 PMCID: PMC4803735 DOI: 10.3389/fimmu.2016.00107] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/08/2016] [Indexed: 01/21/2023] Open
Abstract
Allergic asthma is a chronic inflammatory disease of the airways that is driven by maladaptive T helper 2 (Th2) and Th17 immune responses against harmless, airborne substances. Pulmonary phagocytes represent the first line of defense in the lung where they constantly sense the local environment for potential threats. They comprise two distinct cell types, i.e., macrophages and dendritic cells (DC) that differ in their origins and functions. Alveolar macrophages quickly take up most of the inhaled allergens, yet do not deliver their cargo to naive T cells sampling in draining lymph nodes. In contrast, pulmonary DCs instruct CD4(+) T cells develop into Th2 and Th17 effectors, initiating the maladaptive immune responses toward harmless environmental substances observed in allergic individuals. Unraveling the mechanisms underlying this mistaken identity of harmless, airborne substances by innate immune cells is one of the great challenges in asthma research. The identification of different pulmonary DC subsets, their role in antigen uptake, migration to the draining lymph nodes, and their potential to instruct distinct T cell responses has set the stage to unravel this mystery. However, at this point, a detailed understanding of the spatiotemporal resolution of DC subset localization, allergen uptake, processing, autocrine and paracrine cellular crosstalk, and the humoral factors that define the activation status of DCs is still lacking. In addition to DCs, at least two distinct macrophage populations have been identified in the lung that are either located in the airway/alveolar lumen or in the interstitium. Recent data suggest that such populations can exert either pro- or anti-inflammatory functions. Similar to the DC subsets, detailed insights into the individual roles of alveolar and interstitial macrophages during the different phases of asthma development are still missing. Here, we will provide an update on the current understanding of the origin, localization, and function of the diverse pulmonary antigen-presenting cell subsets, in particular with regard to the development and regulation of allergic asthma. While most data are from mouse models of experimental asthma, we have also included available human data to judge the translational value of the findings obtained in experimental asthma models.
Collapse
Affiliation(s)
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
83
|
Goossens PL, Tournier JN. Crossing of the epithelial barriers by Bacillus anthracis: the Known and the Unknown. Front Microbiol 2015; 6:1122. [PMID: 26500645 PMCID: PMC4598578 DOI: 10.3389/fmicb.2015.01122] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 09/28/2015] [Indexed: 12/15/2022] Open
Abstract
Anthrax, caused by Bacillus anthracis, a Gram-positive spore-forming bacterium, is initiated by the entry of spores into the host body. There are three types of human infection: cutaneous, inhalational, and gastrointestinal. For each form, B. anthracis spores need to cross the cutaneous, respiratory or digestive epithelial barriers, respectively, as a first obligate step to establish infection. Anthrax is a toxi-infection: an association of toxemia and rapidly spreading infection progressing to septicemia. The pathogenicity of Bacillus anthracis mainly depends on two toxins and a capsule. The capsule protects bacilli from the immune system, thus promoting systemic dissemination. The toxins alter host cell signaling, thereby paralyzing the immune response of the host and perturbing the endocrine and endothelial systems. In this review, we will mainly focus on the events and mechanisms leading to crossing of the respiratory epithelial barrier, as the majority of studies have addressed inhalational infection. We will discuss the critical gaps of knowledge that need to be addressed to gain a comprehensive view of the initial steps of inhalational anthrax. We will then discuss the few data available on B. anthracis crossing the cutaneous and digestive epithelia.
Collapse
Affiliation(s)
- Pierre L Goossens
- Pathogénie des Toxi-Infections Bactériennes, Institut Pasteur , Paris, France
| | - Jean-Nicolas Tournier
- Pathogénie des Toxi-Infections Bactériennes, Institut Pasteur , Paris, France ; Unité Interactions Hôte-Agents Pathogènes, Institut de Recherche Biomédicale des Armées , Brétigny-sur-Orge, France ; Ecole du Val-de-Grâce , Paris, France
| |
Collapse
|