1
|
Herb M, Schatz V, Hadrian K, Hos D, Holoborodko B, Jantsch J, Brigo N. Macrophage variants in laboratory research: most are well done, but some are RAW. Front Cell Infect Microbiol 2024; 14:1457323. [PMID: 39445217 PMCID: PMC11496307 DOI: 10.3389/fcimb.2024.1457323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 09/06/2024] [Indexed: 10/25/2024] Open
Abstract
Macrophages play a pivotal role in the innate immune response. While their most characteristic function is phagocytosis, it is important not to solely characterize macrophages by this activity. Their crucial roles in body development, homeostasis, repair, and immune responses against pathogens necessitate a broader understanding. Macrophages exhibit remarkable plasticity, allowing them to modify their functional characteristics in response to the tissue microenvironment (tissue type, presence of pathogens or inflammation, and specific signals from neighboring cells) swiftly. While there is no single defined "macrophage" entity, there is a diverse array of macrophage types because macrophage ontogeny involves the differentiation of progenitor cells into tissue-resident macrophages, as well as the recruitment and differentiation of circulating monocytes in response to tissue-specific cues. In addition, macrophages continuously sense and respond to environmental cues and tissue conditions, adjusting their functional and metabolic states accordingly. Consequently, it is of paramount importance to comprehend the heterogeneous origins and functions of macrophages employed in in vitro studies, as each available in vitro macrophage model is associated with specific sets of strengths and limitations. This review centers its attention on a comprehensive comparison between immortalized mouse macrophage cell lines and primary mouse macrophages. It provides a detailed analysis of the strengths and weaknesses inherent in these in vitro models. Finally, it explores the subtle distinctions between diverse macrophage cell lines, offering insights into numerous factors beyond the model type that can profoundly influence macrophage function.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Valentin Schatz
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Karina Hadrian
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Hos
- Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Bohdan Holoborodko
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg and University of Regensburg, Regensburg, Germany
| | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Natascha Brigo
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
2
|
Bochner BS, O'Sullivan JA, Chang AT, Youngblood BA. Siglecs in allergy and asthma. Mol Aspects Med 2023; 90:101104. [PMID: 35835621 PMCID: PMC10757266 DOI: 10.1016/j.mam.2022.101104] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/28/2022] [Accepted: 07/03/2022] [Indexed: 01/21/2023]
Abstract
The term "allergic diseases" encompasses several common, IgE-mediated conditions that range from being annoying to those that are life-threatening. Available treatments include active avoidance of the instigating allergen and the use of a variety of oral, inhaled, intranasal, intraocular and injected agents. While most individuals with allergies do well with existing therapies, there are still unmet therapeutic needs. Siglecs (sialic acid-binding, immunoglobulin-like lectins) are a family of single-pass transmembrane I-type lectins found on various subsets of cells, especially those of the immune system. All Siglecs have extracellular domains recognizing sialoside ligands, and most contain cytoplasmic domains with inhibitory signaling activity. This review focuses on Siglecs that likely play a role in regulating allergic and asthmatic responses, and how specific Siglecs, expressed on cells such as eosinophils and mast cells, are being targeted for therapeutic benefit.
Collapse
Affiliation(s)
- Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Jeremy A O'Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | |
Collapse
|
3
|
Shah SD, Nayak AP, Sharma P, Villalba DR, Addya S, Huang W, Shapiro P, Kane MA, Deshpande DA. Targeted Inhibition of Select Extracellular Signal-regulated Kinases 1 and 2 Functions Mitigates Pathological Features of Asthma in Mice. Am J Respir Cell Mol Biol 2023; 68:23-38. [PMID: 36067041 PMCID: PMC9817918 DOI: 10.1165/rcmb.2022-0110oc] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/26/2022] [Indexed: 02/05/2023] Open
Abstract
ERK1/2 (extracellular signal-regulated kinases 1 and 2) regulate the activity of various transcription factors that contribute to asthma pathogenesis. Although an attractive drug target, broadly inhibiting ERK1/2 is challenging because of unwanted cellular toxicities. We have identified small molecule inhibitors with a benzenesulfonate scaffold that selectively inhibit ERK1/2-mediated activation of AP-1 (activator protein-1). Herein, we describe the findings of targeting ERK1/2-mediated substrate-specific signaling with the small molecule inhibitor SF-3-030 in a murine model of house dust mite (HDM)-induced asthma. In 8- to 10-week-old BALB/c mice, allergic asthma was established by repeated intranasal HDM (25 μg/mouse) instillation for 3 weeks (5 days/week). A subgroup of mice was prophylactically dosed with 10 mg/kg SF-3-030/DMSO intranasally 30 minutes before the HDM challenge. Following the dosing schedule, mice were evaluated for alterations in airway mechanics, inflammation, and markers of airway remodeling. SF-3-030 treatment significantly attenuated HDM-induced elevation of distinct inflammatory cell types and cytokine concentrations in BAL and IgE concentrations in the lungs. Histopathological analysis of lung tissue sections revealed diminished HDM-induced pleocellular peribronchial inflammation, mucus cell metaplasia, collagen accumulation, thickening of airway smooth muscle mass, and expression of markers of cell proliferation (Ki-67 and cyclin D1) in mice treated with SF-3-030. Furthermore, SF-3-030 treatment attenuated HDM-induced airway hyperresponsiveness in mice. Finally, mechanistic studies using transcriptome and proteome analyses suggest inhibition of HDM-induced genes involved in inflammation, cell proliferation, and tissue remodeling by SF-3-030. These preclinical findings demonstrate that function-selective inhibition of ERK1/2 signaling mitigates multiple features of asthma in a murine model.
Collapse
Affiliation(s)
- Sushrut D. Shah
- Center for Translational Medicine, Jane and Leonard Korman Lung Center, and
| | - Ajay P. Nayak
- Center for Translational Medicine, Jane and Leonard Korman Lung Center, and
| | - Pawan Sharma
- Center for Translational Medicine, Jane and Leonard Korman Lung Center, and
| | | | - Sankar Addya
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania; and
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, Maryland
| | - Paul Shapiro
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, Maryland
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Baltimore, Maryland
| | | |
Collapse
|
4
|
Differential Activity of Human Leukocyte Extract on Systemic Immune Response and Cyst Growth in Mice with Echinococcus Multilocularis Infection After Oral, Subcutaneous and Intraperitoneal Routes of Administration. Helminthologia 2022; 59:341-356. [PMID: 36875680 PMCID: PMC9979067 DOI: 10.2478/helm-2022-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/04/2022] [Indexed: 02/05/2023] Open
Abstract
Alveolar echinococcosis (AE) caused by the larval stage of Echinococcus multilocularis is serious parasitic diseases associated with the host´s immunosuppression. The effects of human non-immune dialyzable leukocyte extract (DLE) on immune cells in blood and spleen and parasitic cysts weight in Balb/c mice after oral (PO), subcutaneous (SC) and intraperitoneal administration (IP) were compared. The reduction in cysts weight (p < 0.01) was recorded after PO route, whereas moderate reduction was found after SC and IP routes. The elevation of lymphoid populations in blood and spleen was found after PO administration (p < 0.01) in parallel with reduced myeloid population. Infection-elicited decline in B220+B cells was partially abolished by PO route, but DLE routes did not influence the CD3+ T cells. The proportions of CD3+CD4+Th lymphocytes were moderately upregulated, whereas CD3+CD8+Tc populations were reduced after all DLE routes (p < 0.01). PO administration increased CD11b+MHCIIhigh blood monocytes, CD11b-SigleF+ cell, but not CD11b+Si-glecF+ eosinophils in the blood, stimulated after SC and IP routes. DLE induced downregulation of NO production by LPS-stimulated adherent splenocytes ex vivo. Con A-triggered T lymphocyte proliferation was associated with the elevated IFN-γ production and transcription factor Tbet mRNA expression. The alleviation of Th2 (IL-4) and Treg (TGF-β) cytokine production by lymphocytes ex vivo paralleled with downregulation of gene transcription for cytokines, GATA and FoxP3. Reduction of myeloid cells with suppressive activity was found. The SC and IP routes affected partially the cysts weights, diminished significantly gene transcription, NO levels and Th2 and Treg cytokines production. Results showed that PO route of DLE administration was the most effective in ameliorating immunosuppression via stimulation of Th1 type, reducing Th2 and Treg type of immunity and CD3+CD8+Tc lymphocytes in the blood and spleens during E. multilocularis infection in mice.
Collapse
|
5
|
Mabille D, Dirkx L, Thys S, Vermeersch M, Montenye D, Govaerts M, Hendrickx S, Takac P, Van Weyenbergh J, Pintelon I, Delputte P, Maes L, Pérez-Morga D, Timmermans JP, Caljon G. Impact of pulmonary African trypanosomes on the immunology and function of the lung. Nat Commun 2022; 13:7083. [PMID: 36400767 PMCID: PMC9674601 DOI: 10.1038/s41467-022-34757-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 11/04/2022] [Indexed: 11/19/2022] Open
Abstract
Approximately 20% of sleeping sickness patients exhibit respiratory complications, however, with a largely unknown role of the parasite. Here we show that tsetse fly-transmitted Trypanosoma brucei parasites rapidly and permanently colonize the lungs and occupy the extravascular spaces surrounding the blood vessels of the alveoli and bronchi. They are present as nests of multiplying parasites exhibiting close interactions with collagen and active secretion of extracellular vesicles. The local immune response shows a substantial increase of monocytes, macrophages, dendritic cells and γδ and activated αβ T cells and a later influx of neutrophils. Interestingly, parasite presence results in a significant reduction of B cells, eosinophils and natural killer cells. T. brucei infected mice show no infection-associated pulmonary dysfunction, mirroring the limited pulmonary clinical complications during sleeping sickness. However, the substantial reduction of the various immune cells may render individuals more susceptible to opportunistic infections, as evident by a co-infection experiment with respiratory syncytial virus. Collectively, these observations provide insights into a largely overlooked target organ, and may trigger new diagnostic and supportive therapeutic approaches for sleeping sickness.
Collapse
Affiliation(s)
- Dorien Mabille
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Laura Dirkx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Sofie Thys
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Marjorie Vermeersch
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Daniel Montenye
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Matthias Govaerts
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Sarah Hendrickx
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Peter Takac
- Institute of Zoology, Slovak Academy of Sciences, 84506, Bratislava, Slovakia
- Scientica, Ltd., 83106, Bratislava, Slovakia
| | - Johan Van Weyenbergh
- Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute of Medical Research, KU Leuven, Leuven, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Peter Delputte
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Louis Maes
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium
| | - David Pérez-Morga
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Gosselies, Belgium
- Laboratory of Molecular Parasitology, IBMM, Université libre de Bruxelles, Gosselies, Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| | - Guy Caljon
- Laboratory of Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp, Wilrijk, Belgium.
| |
Collapse
|
6
|
Chudakov DB, Konovalova MV, Kashirina EI, Kotsareva OD, Shevchenko MA, Tsaregorodtseva DS, Fattakhova GV. DEPs Induce Local Ige Class Switching Independent of Their Ability to Stimulate iBALT de Novo Formation. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:13063. [PMID: 36293642 PMCID: PMC9603618 DOI: 10.3390/ijerph192013063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/04/2022] [Accepted: 10/08/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Diesel exhaust particles (DEPs) are leading to a general increase in atopic diseases worldwide. However, it is still unknown whether DEPs induce systemic B-cell IgE class switching in secondary lymphoid organs or locally in the lungs in inducible bronchus-associated lymphoid tissue (iBALT). The aim of this work was to identify the exact site of DEP-mediated B-cell IgE class switching and pro-allergic antibody production. METHODS We immunized BALB/c mice with different OVA doses (0.3 and 30 µg) intranasally in the presence and absence of two types of DEPs, SRM1650B and SRM2786. We used low (30 µg) and high (150 µg) DEP doses. RESULTS Only a high DEP dose induced IgE production, regardless of the particle type. Local IgE class switching was stimulated upon treatment with both types of particles with both low and high OVA doses. Despite the similar ability of the two standard DEPs to stimulate IgE production, their ability to induce iBALT formation and growth was markedly different upon co-administration with low OVA doses. CONCLUSIONS DEP-induced local IgE class switching takes place in preexisting iBALTs independent of de novo iBALT formation, at least in the case of SRM1650B co-administered with low OVA doses.
Collapse
Affiliation(s)
- Dmitrii Borisovich Chudakov
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya Str., Moscow 117997, Russia
| | - Mariya Vladimirovna Konovalova
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya Str., Moscow 117997, Russia
| | - Elena Igorevna Kashirina
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya Str., Moscow 117997, Russia
| | - Olga Dmitrievna Kotsareva
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya Str., Moscow 117997, Russia
| | - Marina Alexandrovna Shevchenko
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya Str., Moscow 117997, Russia
| | - Daria Sergeevna Tsaregorodtseva
- Faculty of Medical Biology, Sechenov First Moscow State Medical University, 2 Bolshaya Pirogovskaya Str., Moscow 1194535, Russia
| | - Gulnar Vaisovna Fattakhova
- Laboratory of Cell Interactions, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 16/10 Miklukho-Maklaya Str., Moscow 117997, Russia
| |
Collapse
|
7
|
Human and Mouse Eosinophils Differ in Their Ability to Biosynthesize Eicosanoids, Docosanoids, the Endocannabinoid 2-Arachidonoyl-glycerol and Its Congeners. Cells 2022; 11:cells11010141. [PMID: 35011703 PMCID: PMC8750928 DOI: 10.3390/cells11010141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 12/13/2022] Open
Abstract
High eosinophil (EOS) counts are a key feature of eosinophilic asthma. EOS notably affect asthmatic response by generating several lipid mediators. Mice have been utilized in hopes of defining new pharmacological targets to treat asthma. However, many pinpointed targets in mice did not translate into clinics, underscoring that key differences exist between the two species. In this study, we compared the ability of human (h) and mouse (m) EOS to biosynthesize key bioactive lipids derived from arachidonic acid (AA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). hEOS were isolated from the blood of healthy subjects and mild asthmatics, while mEOSs were differentiated from the bone marrow. EOSs were treated with fatty acids and lipid mediator biosynthesis assessed by LC-MS/MS. We found that hEOS biosynthesized leukotriene (LT) C4 and LTB4 in a 5:1 ratio while mEOS almost exclusively biosynthesized LTB4. The biosynthesis of the 15-lipoxygenase (LO) metabolites 15-HETE and 12-HETE also differed, with a 15-HETE:12-HETE ratio of 6.3 for hEOS and 0.727 for mEOS. EOS biosynthesized some specialized pro-resolving mediators, and the levels from mEOS were 9-times higher than those of hEOS. In contrast, hEOS produced important amounts of the endocannabinoid 2-arachidonoyl-glycerol (2-AG) and its congeners from EPA and DHA, a biosynthetic pathway that was up to ~100-fold less prominent in mEOS. Our data show that hEOS and mEOS biosynthesize the same lipid mediators but in different amounts. Compared to asthmatics, mouse models likely have an amplified involvement of LTB4 and specialized pro-resolving mediators and a diminished impact of the endocannabinoid 2-arachidonoyl-glycerol and its congeners.
Collapse
|
8
|
Jeong BM, Walker MT, Rodriguez R, Coden ME, Nagasaka R, Doan TC, Politanska Y, Abdala-Valencia H, Berdnikovs S. More than neutrophils: Lin(+)Ly6G(+)IL-5Rα(+) multipotent myeloid cells (MMCs) are dominant in normal murine bone marrow and retain capacity to differentiate into eosinophils and monocytes. J Leukoc Biol 2022; 111:113-122. [PMID: 33857341 PMCID: PMC10080214 DOI: 10.1002/jlb.1ab0519-170rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Bone marrow is a hematopoietic site harboring multiple populations of myeloid cells in different stages of differentiation. Murine bone marrow eosinophils are traditionally identified by Siglec-F(+) staining using flow cytometry, whereas neutrophils are characterized by Ly6G(+) expression. However, using flow cytometry to characterize bone marrow hematopoietic cells in wild-type mice, we found substantial gray areas in identification of these cells. Siglec-F(+) mature eosinophil population constituted only a minority of bone marrow Lin(+)CD45(+) pool (5%). A substantial population of Siglec-F(-) cells was double positive for neutrophil marker Ly6G and eosinophil lineage marker, IL-5Rα. This granulocyte population with mixed neutrophil and eosinophil characteristics is typically attributable to neutrophil pool based on neutral granule staining and expression of Ly6G and myeloid peroxidase. It is distinct from Lineage(-) myeloid progenitors or Siglec-F(+)Ly6G(+) maturing eosinophil precursors, and can be accurately identified by Lineage(+) staining and positive expression of markers IL-5Rα and Ly6G. At 15-50% of all CD45(+) hematopoietic cells in adult mice (percentage varies by sex and age), this is a surprisingly dominant population, which increases with age in both male and female mice. RNA-seq characterization of these cells revealed a complex immune profile and the capacity to secrete constituents of the extracellular matrix. When sorted from bone marrow, these resident cells had neutrophilic phenotype but readily acquired all characteristics of eosinophils when cultured with G-CSF or IL-5, including expression of Siglec-F and granular proteins (Epx, Mbp). Surprisingly, these cells were also able to differentiate into Ly6C(+) monocytes when cultured with M-CSF. Herein described is the discovery of an unexpected hematopoietic flexibility of a dominant population of multipotent myeloid cells, typically categorized as neutrophils, but with the previously unknown plasticity to contribute to mature pools of eosinophils and monocytes.
Collapse
Affiliation(s)
- Brian M. Jeong
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew T. Walker
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Raul Rodriguez
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mackenzie E. Coden
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Reina Nagasaka
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ton C. Doan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yuliya Politanska
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sergejs Berdnikovs
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
9
|
Broberg L, González-Cano P, Arsic N, Popowych Y, Griebel PJ. Isolation and characterization of eosinophils in bovine blood and small intestine. Vet Immunol Immunopathol 2021; 242:110352. [PMID: 34773748 DOI: 10.1016/j.vetimm.2021.110352] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/01/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022]
Abstract
An effective method to isolate functional eosinophils from blood and tissues is required to analyze the multiple roles eosinophils play in innate immunity and tissue homeostasis. Highspeed cell sorting was used to isolate bovine eosinophils from blood polymorphonuclear (PMN) cells and from small intestine intraepithelial leukocytes. Eosinophils and neutrophils were purified from bovine blood with highspeed cell sorting after gating on autofluorescence (FL1) high and low PMN subpopulations. Highspeed sorting of intestinal eosinophils was accomplished by using a combination of positive (CD45+, CD11cLow, side scatterHigh) and negative (CD3-) selection parameters. Eosinophils sorted from blood PMNs were 88.6 ± 5.8 % (mean + 1 SD; n = 4) pure and yielded significantly (p < 0.05) more RNA than purified neutrophils. Analysis of Toll-like receptor (TLR) gene expression and TLR ligand-induced pro-inflammatory cytokine (IL-1, IL-6, IL-8, and TNFα) gene expression demonstrated significant (p < 0.01) functional differences between blood eosinophils and neutrophils. Eosinophils varied between 14.7 % to 29.3 % of CD45+ IELs and purity of sorted intestinal eosinophils was 95 + 3.5 % (mean + 1SD; n = 5). A comparison of mucosal and blood eosinophils revealed significant (p < 0.01) differences in TLR gene expression, supporting the hypothesis that functionally distinct eosinophil populations are present in blood and tissues. In conclusion, highspeed cell sorting provides an effective method to isolate viable eosinophils from blood and tissues that can then be used for transcriptome analyses and in vitro function assays.
Collapse
Affiliation(s)
- Lindsey Broberg
- Vaccine and Infectious Disease Organization, 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada
| | - Patricia González-Cano
- Vaccine and Infectious Disease Organization, 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada
| | - Natasa Arsic
- Vaccine and Infectious Disease Organization, 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada
| | - Yurij Popowych
- Vaccine and Infectious Disease Organization, 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada
| | - Philip J Griebel
- Vaccine and Infectious Disease Organization, 120 Veterinary Road, University of Saskatchewan, Saskatoon, SK, S7N 5E3, Canada; School of Public Health, University of Saskatchewan, Saskatoon, SK, S7N 2Z4, Canada.
| |
Collapse
|
10
|
Daneshmandi S, Cassel T, Higashi RM, Fan TWM, Seth P. 6-Phosphogluconate dehydrogenase (6PGD), a key checkpoint in reprogramming of regulatory T cells metabolism and function. eLife 2021; 10:e67476. [PMID: 34709178 PMCID: PMC8553335 DOI: 10.7554/elife.67476] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 10/17/2021] [Indexed: 12/29/2022] Open
Abstract
Cellular metabolism has key roles in T cells differentiation and function. CD4+ T helper-1 (Th1), Th2, and Th17 subsets are highly glycolytic while regulatory T cells (Tregs) use glucose during expansion but rely on fatty acid oxidation for function. Upon uptake, glucose can enter pentose phosphate pathway (PPP) or be used in glycolysis. Here, we showed that blocking 6-phosphogluconate dehydrogenase (6PGD) in the oxidative PPP resulted in substantial reduction of Tregs suppressive function and shifts toward Th1, Th2, and Th17 phenotypes which led to the development of fetal inflammatory disorder in mice model. These in turn improved anti-tumor responses and worsened the outcomes of colitis model. Metabolically, 6PGD blocked Tregs showed improved glycolysis and enhanced non-oxidative PPP to support nucleotide biosynthesis. These results uncover critical role of 6PGD in modulating Tregs plasticity and function, which qualifies it as a novel metabolic checkpoint for immunotherapy applications.
Collapse
Affiliation(s)
- Saeed Daneshmandi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
- Division of Interdisciplinary Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
| | - Teresa Cassel
- Center for Environmental and Systems Biochemistry, University of KentuckyLexingtonUnited States
| | - Richard M Higashi
- Center for Environmental and Systems Biochemistry, University of KentuckyLexingtonUnited States
| | - Teresa W-M Fan
- Center for Environmental and Systems Biochemistry, University of KentuckyLexingtonUnited States
| | - Pankaj Seth
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
- Division of Interdisciplinary Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
11
|
Vélez-del-Burgo A, Sánchez P, Suñen E, Martínez J, Postigo I. Purified Native and Recombinant Major Alternaria alternata Allergen (Alt a 1) Induces Allergic Asthma in the Murine Model. J Fungi (Basel) 2021; 7:jof7110896. [PMID: 34829186 PMCID: PMC8624818 DOI: 10.3390/jof7110896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/23/2022] Open
Abstract
Aeroallergens such us the spores of Alternaria alternata are described as the most important agents associated with respiratory allergies and severe asthma. Various experimental models of asthma have been developed using A. alternata extracts to study the pathogenesis of asthma, establishing the main parameters that trigger the asthmatic response. In this study, we describe a mouse model of asthma induced only by Alt a 1. To induce the allergic response, mice were challenged intranasally with the major allergen of A. alternata, Alt a 1. The presence of eosinophils in the lungs, elevated concentrations of Th2 family cytokines, lymphocyte proliferation and elevated IgE total serum levels indicated that the sensitisation and challenge with Alt a 1 induced the development of airway inflammation. Histological studies showed an eosinophilic cellular infiltrate in the lung tissue of mice instilled with Alt a 1. We demonstrate that Alt a 1 alone is capable of inducing a lung inflammatory response with an increase in IgE serum levels mimicking the allergic asthma immunoresponse when it is administered into BALB/c mice. This model will allow the evaluation of the immunoregulatory or immunotolerant capacity of several molecules that can be used in targeted immunotherapy for fungal allergic asthma.
Collapse
|
12
|
Mair I, Wolfenden A, Lowe AE, Bennett A, Muir A, Smith H, Fenn J, Bradley JE, Else KJ. A lesson from the wild: The natural state of eosinophils is Ly6G hi. Immunology 2021; 164:766-776. [PMID: 34486729 PMCID: PMC8561109 DOI: 10.1111/imm.13413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/29/2021] [Accepted: 09/02/2021] [Indexed: 12/11/2022] Open
Abstract
With a long history of promoting pathological inflammation, eosinophils are now emerging as important regulatory cells. Yet, findings from controlled laboratory experiments so far lack translation to animals, including humans, in their natural environment. In order to appreciate the breadth of eosinophil phenotype under non‐laboratory, uncontrolled conditions, we exploit a free‐living population of the model organism Mus musculus domesticus. Eosinophils were present at significantly higher proportions in the spleen and bone marrow of wild mice compared with laboratory mice. Strikingly, the majority of eosinophils of wild mice exhibited a unique Ly6Ghi phenotype seldom described in laboratory literature. Ly6G expression correlated with activation status in spleen and bone marrow, but not peritoneal exudate cells, and is therefore likely not an activation marker per se. Intermediate Ly6G expression was transiently induced in a small proportion of eosinophils from C57BL/6 laboratory mice during acute infection with the whipworm Trichuris muris, but not during low‐dose chronic infection, which better represents parasite exposure in the wild. We conclude that the natural state of the eosinophil is not adequately reflected in the standard laboratory mouse, which compromises our attempts to dissect their functional relevance. Our findings emphasize the importance of studying the immune system in its natural context – alongside more mechanistic laboratory experiments – in order to capture the entirety of immune phenotypes and functions.
Collapse
Affiliation(s)
- Iris Mair
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew Wolfenden
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Ann E Lowe
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Alex Bennett
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew Muir
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Hannah Smith
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jonathan Fenn
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Kathryn J Else
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
13
|
Bounds K, Colmer-Hamood JA, Myntti M, Jeter RM, Hamood AN. The influence of a biofilm-dispersing wound gel on the wound healing process. Int Wound J 2021; 19:553-572. [PMID: 34263993 PMCID: PMC8874046 DOI: 10.1111/iwj.13653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/14/2021] [Accepted: 06/21/2021] [Indexed: 01/02/2023] Open
Abstract
Topical antimicrobials that reduce the bacterial bioburden within a chronically‐infected wound may have helpful or harmful effects on the healing process. We used murine models of full‐thickness skin wounds to determine the effects of the novel biofilm‐dispersing wound gel (BDWG) and its gel base on the healing of uninfected wounds. The rate of wound closure over 19 days was comparable among the BDWG‐treated (BT) wounds and the controls. Compared with the controls, histology of the BT wounds showed formation of a stable blood clot at day 1, more neovascularisation and reepithelialisation at day 3, and more organised healing at day 7. Fluorescence‐activated cell sorting analysis showed a lower percentage of neutrophils in wounded tissues of the BT group at days 1 and 3, and significantly more M2 macrophages at day 3. Levels of proinflammatory cytokines and chemokines were increased over the uninjured baseline within the wounds of all treatment groups but the levels were significantly lower in the BT group at day 1, modulating the inflammatory response. Our results suggest that BDWG does not interfere with the wound healing process and may enhance it by lowering inflammation and allowing transition to the proliferative stage of wound healing by day 3.
Collapse
Affiliation(s)
- Kayla Bounds
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA.,Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Jane A Colmer-Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Matthew Myntti
- Research and Development, Next Science LLC, Jacksonville, Florida, USA
| | - Randall M Jeter
- Department of Biological Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Abdul N Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA.,Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| |
Collapse
|
14
|
Wiley MB, Bobardt SD, Nordgren TM, Nair MG, DiPatrizio NV. Cannabinoid Receptor Subtype-1 Regulates Allergic Airway Eosinophilia During Lung Helminth Infection. Cannabis Cannabinoid Res 2021; 6:242-252. [PMID: 33998896 PMCID: PMC8217601 DOI: 10.1089/can.2020.0167] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Introduction: Over 1 billion humans carry infectious helminth parasites that can lead to chronic comorbidities such as anemia and growth retardation in children. Helminths induce a T-helper type 2 (Th2) immune response in the host and can cause severe tissue damage and fibrosis if chronic. We recently reported that mice infected with the soil-transmitted helminth, Nippostrongylus brasiliensis, displayed elevated levels of endocannabinoids (eCBs) in the lung and intestine. eCBs are lipid-signaling molecules that control inflammation; however, their function in infection is not well defined. Materials and Methods: A combination of pharmacological approaches and genetic mouse models was used to investigate roles for the eCB system in inflammatory responses and lung injury in mice during parasitic infection with N. brasiliensis. Results: Hemorrhaging of lung tissue in mice infected with N. brasiliensis was exacerbated by inhibiting peripheral cannabinoid receptor subtype-1 (CB1Rs) with the peripherally restricted CB1R antagonist, AM6545. In addition, these mice exhibited an increase in nonfunctional alveolar space and prolonged airway eosinophilia compared to vehicle-treated infected mice. In contrast to mice treated with AM6545, infected cannabinoid receptor subtype-2-null mice (Cnr2-/-) did not display any changes in these parameters compared to wild-type mice. Conclusions: Roles for the eCB system in Th2 immune responses are not well understood; however, increases in its activity in response to infection suggest an immunomodulatory role. Moreover, these findings suggest a role for eCB signaling at CB1Rs but not cannabinoid receptor subtypes-2 in the resolution of Th2 inflammatory responses, which become host destructive over time.
Collapse
Affiliation(s)
- Mark B. Wiley
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Sarah D. Bobardt
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Tara M. Nordgren
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Meera G. Nair
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| | - Nicholas V. DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, California, USA
| |
Collapse
|
15
|
Nutritional immunity: the impact of metals on lung immune cells and the airway microbiome during chronic respiratory disease. Respir Res 2021; 22:133. [PMID: 33926483 PMCID: PMC8082489 DOI: 10.1186/s12931-021-01722-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022] Open
Abstract
Nutritional immunity is the sequestration of bioavailable trace metals such as iron, zinc and copper by the host to limit pathogenicity by invading microorganisms. As one of the most conserved activities of the innate immune system, limiting the availability of free trace metals by cells of the immune system serves not only to conceal these vital nutrients from invading bacteria but also operates to tightly regulate host immune cell responses and function. In the setting of chronic lung disease, the regulation of trace metals by the host is often disrupted, leading to the altered availability of these nutrients to commensal and invading opportunistic pathogenic microbes. Similarly, alterations in the uptake, secretion, turnover and redox activity of these vitally important metals has significant repercussions for immune cell function including the response to and resolution of infection. This review will discuss the intricate role of nutritional immunity in host immune cells of the lung and how changes in this fundamental process as a result of chronic lung disease may alter the airway microbiome, disease progression and the response to infection.
Collapse
|
16
|
Zhu C, Weng QY, Zhou LR, Cao C, Li F, Wu YF, Wu YP, Li M, Hu Y, Shen JX, Xiong XF, Lan F, Xia LX, Zhang B, Zhang H, Huang M, Ying SM, Shen HH, Chen ZH, Li W. Homeostatic and early-recruited CD101 - eosinophils suppress endotoxin-induced acute lung injury. Eur Respir J 2020; 56:13993003.02354-2019. [PMID: 32527738 DOI: 10.1183/13993003.02354-2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/01/2020] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Acute lung injury (ALI) is a fatal but undertreated condition with severe neutrophilic inflammation, although little is known about the functions of eosinophils in the pathogenesis of ALI. Our objectives were to investigate the roles and molecular mechanisms of eosinophils in ALI. METHODS Pulmonary eosinophils were identified by flow cytometry. Mice with abundant or deficient eosinophils were used. Cellularity of eosinophils and neutrophils in bronchoalveolar lavage fluid, inflammatory assessment, and survival rate were determined. Human samples were also used for validating experimental results. RESULTS Blood eosinophils were increased in surviving patients with acute respiratory distress syndrome (ARDS) independent of corticosteroid usage. There existed homeostatic eosinophils in lung parenchyma in mice and these homeostatic eosinophils, originating from the bone marrow, were predominantly CD101-. More CD101- eosinophils could be recruited earlier than lipopolysaccharide (LPS)-initiated neutrophilic inflammation. Loss of eosinophils augmented LPS-induced pulmonary injury. Homeostatic CD101- eosinophils ameliorated, while allergic CD101+ eosinophils exacerbated, the neutrophilic inflammation induced by LPS. Likewise, CD101 expression in eosinophils from ARDS patients did not differ from healthy subjects. Mechanistically, CD101- eosinophils exhibited higher levels of Alox15 and Protectin D1. Administration of Protectin D1 isomer attenuated the neutrophilic inflammation. CONCLUSIONS Collectively, our findings identify an uncovered function of native CD101- eosinophils in suppressing neutrophilic lung inflammation and suggest a potential therapeutic target for ALI.
Collapse
Affiliation(s)
- Chen Zhu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,These authors contributed equally to this article
| | - Qing-Yu Weng
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,These authors contributed equally to this article
| | - Ling-Ren Zhou
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,These authors contributed equally to this article
| | - Chao Cao
- Dept of Respiratory Medicine, Ningbo First Hospital, Ningbo, China
| | - Fei Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yin-Fang Wu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yan-Ping Wu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Miao Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yue Hu
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jia-Xin Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xue-Fang Xiong
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,Dept of Respiratory Medicine, The Central Hospital of Lishui City, Lishui, China
| | - Fen Lan
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Li-Xia Xia
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Zhang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Zhang
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Man Huang
- Dept of Central Intensive Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Song-Min Ying
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hua-Hao Shen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,State Key Lab for Respiratory Diseases, Guangzhou, China.,These authors contributed equally to this article as lead authors and supervised the work
| | - Zhi-Hua Chen
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,These authors contributed equally to this article as lead authors and supervised the work
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Dept of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.,These authors contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
17
|
Wisnewski AV, Liu J, Redlich CA. Analysis of Lung Gene Expression Reveals a Role for Cl - Channels in Diisocyanate-induced Airway Eosinophilia in a Mouse Model of Asthma Pathology. Am J Respir Cell Mol Biol 2020; 63:25-35. [PMID: 32101465 PMCID: PMC7328250 DOI: 10.1165/rcmb.2019-0400oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 02/25/2020] [Indexed: 12/23/2022] Open
Abstract
Diisocyanates are well-recognized causes of asthma. However, sensitized workers frequently lack diisocyanate-specific IgE, which complicates diagnosis and suggests the disease involves IgE-independent mechanisms. We used a mouse model of methylene diphenyl diisocyanate (MDI) asthma to identify biological pathways that may contribute to asthma pathogenesis. MDI sensitization and respiratory tract exposure were performed in Balb/c, transgenic B-cell (e.g., IgE)-deficient mice and a genetic background (C57BL/6)-matched strain. Eosinophils in airway fluid were quantitated by flow cytometry. Lung tissue gene expression was assessed using whole-genome mRNA microarrays. Informatic software was used to identify biological pathways affected by respiratory tract exposure and potential targets for disease intervention. Airway eosinophilia and changes (>1.5-fold; P value < 0.05) in expression of 192 genes occurred in all three mouse strains tested, with enrichment in chemokines and a pattern associated with alternatively activated monocytes/macrophages. CLCA1 (calcium-activated chloride channel regulator 1) was the most upregulated gene transcript (>100-fold) in all exposed mouse lungs versus controls, followed closely by SLC26A4, another transcript involved in Cl- conductance. Crofelemer, a U.S. Food and Drug Administration-approved Cl- channel inhibitor, reduced MDI exposure induction of airway eosinophilia, mucus, CLCA1, and other asthma-associated gene transcripts. Expression changes in a core set of genes occurs independent of IgE in a mouse model of chemical-induced airway eosinophilia. In addition to chemokines and alternatively activated monocytes/macrophages, the data suggest a crucial role for Cl- channels in diisocyanate asthma pathology and as a possible target for intervention.
Collapse
Affiliation(s)
- Adam V Wisnewski
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jian Liu
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Carrie A Redlich
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
18
|
Knuplez E, Krier-Burris R, Cao Y, Marsche G, O’Sullivan J, Bochner BS. Frontline Science: Superior mouse eosinophil depletion in vivo targeting transgenic Siglec-8 instead of endogenous Siglec-F: Mechanisms and pitfalls. J Leukoc Biol 2020; 108:43-58. [PMID: 32134149 PMCID: PMC7585130 DOI: 10.1002/jlb.3hi0120-381r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 01/02/2023] Open
Abstract
Eosinophils are important multifunctional granulocytes. When studying eosinophil function and its contribution to diseases, mouse models are often used. Mouse eosinophils selectively express sialic acid-binding immunoglobulin-like lectin (Siglec)-F. Its closest functional paralog on human eosinophils is Siglec-8. These Siglecs are being used to target eosinophils when exploring their mechanistic roles in disease and for potential therapeutic benefit. In order to facilitate preclinical studies of human Siglec-8, we developed transgenic mouse strains expressing human Siglec-8 only on the surface of eosinophils with or without endogenous Siglec-F and have begun characterizing various cellular functions in vitro and in vivo. Eosinophils from Siglec-8+ mice, with or without Siglec-F, responded to Siglec-8 antibody engagement in vitro by up-regulating surface CD11b, whereas Siglec-F antibody had no such effect. Engagement of Siglec-F or Siglec-8 with respective antibodies in vitro resulted in only modest increases in cell death. Administration of rat Siglec-F antibodies to mice led to a significant decrease in Siglec-F surface expression on eosinophils due to internalization, and thus appeared to decrease eosinophil numbers based on Siglec-F+ cells, but with proper gaiting strategies did not in fact result in significant eosinophil depletion. In marked contrast, administration of mouse Siglec-8 antibodies rapidly and effectively depleted eosinophils from blood and spleens of mice, but an F(ab')2 version did not, indicating an Fc-mediated mechanism for eosinophil depletion in vivo. Siglec-8 expressing mice with or without endogenous Siglec-F will be useful to study Siglec-8-based therapeutics, and may be a preferred approach when acute or chronic eosinophil depletion is needed.
Collapse
Affiliation(s)
- Eva Knuplez
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Rebecca Krier-Burris
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yun Cao
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Gunther Marsche
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Jeremy O’Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bruce S. Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
19
|
Matsui M, Nagakubo D, Satooka H, Hirata T. A novel Siglec-F + neutrophil subset in the mouse nasal mucosa exhibits an activated phenotype and is increased in an allergic rhinitis model. Biochem Biophys Res Commun 2020; 526:599-606. [PMID: 32247614 DOI: 10.1016/j.bbrc.2020.03.122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 03/20/2020] [Indexed: 11/17/2022]
Abstract
Neutrophils are important phagocytic cells for host defense against pathogens. They are rapidly recruited to the site of infection, release antimicrobial peptides and cytokines, and engulf and kill microbes. Neutrophils also accumulate in allergic inflammatory sites. Here we characterized neutrophil accumulation in the nasal mucosa using a mouse model of allergic rhinitis, in which mice were sensitized by intraperitoneal injection of ovalbumin (OVA) and then challenged by intranasal administration of OVA or PBS. In the nasal mucosa of both PBS- and OVA-challenged mice, we found a cell subset expressing the eosinophil marker Siglec-F in the Ly-6G+ neutrophil population. Morphological analysis of the sorted Ly-6G+Siglec-F+ cells revealed that they were devoid of eosinophilic granules in the cytosol and were apparently neutrophils, but compared to conventional Ly-6G+Siglec-F- neutrophils, they had a more lobulated, "botryoid" nucleus. Siglec-F+ neutrophils were barely found in the nasopharynx-associated lymphoid tissue, cervical lymph nodes, the spleen, or blood. Both Siglec-F+ neutrophils and conventional neutrophils showed increased numbers in the nasal mucosa of OVA-challenged mice. Compared to conventional Siglec-F- neutrophils, Siglec-F+ neutrophils exhibited an activated phenotype and enhanced effector functions. Taken together, our findings identify Siglec-F+ neutrophils as a novel neutrophil subset with an activated phenotype that resides specifically in the nasal mucosa.
Collapse
Affiliation(s)
- Makoto Matsui
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| | - Daisuke Nagakubo
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| | - Hiroki Satooka
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| | - Takako Hirata
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| |
Collapse
|
20
|
Beckert H, Meyer-Martin H, Buhl R, Taube C, Reuter S. Single and Synergistic Effects of Type 2 Cytokines on Eosinophils and Asthma Hallmarks. THE JOURNAL OF IMMUNOLOGY 2019; 204:550-558. [PMID: 31862712 DOI: 10.4049/jimmunol.1901116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/19/2019] [Indexed: 12/15/2022]
Abstract
The type 2 cytokines IL-5, IL-13, and IL-4 play an important role in the induction and progression of asthma. According to the Global Initiative for Asthma guidelines, blood eosinophil numbers are one marker that helps to guide treatment decisions in patients suffering from severe forms of asthma. Effects of type 2 cytokines were analyzed, alone or in combination, on eosinophils in blood and other compartments and on the development of asthma symptoms. C57BL/6 mice received a single intranasal application of equimolar amounts of IL-5, IL-13, and IL-4, alone or in combination. Numbers, activation state, and migratory behavior of eosinophils in bone marrow (BM), blood, lung, and bronchoalveolar lavage as well as airway hyperresponsiveness and goblet cell metaplasia were evaluated. Only IL-13 was associated with airway eosinophilia, development of airway hyperresponsiveness, and goblet cell metaplasia, without any synergistic effects. IL-5 increased the number of eosinophils in BM and lung tissue but failed to affect structural changes. IL-4 had similar, but weaker, effects to IL-13. Cytokine combinations synergistically affected eosinophils but failed to enhance IL-13-driven effects on lung function or goblet cell metaplasia. IL-5 and IL-13 markedly increased eosinophil numbers locally in lung and airways and distally in blood and BM, whereas IL-5 and IL-4 only increased eosinophils in lung and BM. IL-13 together with IL-4 failed to demonstrate any synergistic effect. These insights into single and combined effects of type 2 cytokines on disease-driving mechanisms could improve understanding of the impact and effectiveness of new therapies in asthma.
Collapse
Affiliation(s)
- Hendrik Beckert
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, 45239 Essen, Germany; and
| | - Helen Meyer-Martin
- Department of Pulmonary Medicine, III, Medical Clinic, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Roland Buhl
- Department of Pulmonary Medicine, III, Medical Clinic, Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany
| | - Christian Taube
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, 45239 Essen, Germany; and
| | - Sebastian Reuter
- Department of Pulmonary Medicine, University Hospital Essen-Ruhrlandklinik, 45239 Essen, Germany; and
| |
Collapse
|
21
|
Schneider-Ohrum K, Snell Bennett A, Rajani GM, Hostetler L, Maynard SK, Lazzaro M, Cheng LI, O'Day T, Cayatte C. CD4 + T Cells Drive Lung Disease Enhancement Induced by Immunization with Suboptimal Doses of Respiratory Syncytial Virus Fusion Protein in the Mouse Model. J Virol 2019; 93:e00695-19. [PMID: 31092578 PMCID: PMC6639276 DOI: 10.1128/jvi.00695-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection of seronegative children previously immunized with formalin-inactivated (FI) RSV has been associated with serious enhanced respiratory disease (ERD). The phenomenon was reproduced in the cotton rat and the mouse, and both preclinical models have been routinely used to evaluate the safety of new RSV vaccine candidates. More recently, we demonstrated that immunizations with suboptimal doses of the RSV fusion (F) antigen, in its post- or prefusion conformation, and in the presence of a Th1-biasing adjuvant, unexpectedly led to ERD in the cotton rat model. To assess if those observations are specific to the cotton rat and to elucidate the mechanism by which vaccination with low antigen doses can drive ERD post-RSV challenge, we evaluated RSV post-F antigen dose de-escalation in BALB/c mice in the presence of a Th1-biasing adjuvant. While decreasing antigen doses, we observed an increase in lung inflammation associated with an upregulation of proinflammatory cytokines. The amplitude of the lung histopathology was comparable to that of FI-RSV-induced ERD, confirming the observations made in the cotton rat. Importantly, depletion of CD4+ T cells prior to viral challenge completely abrogated ERD, preventing proinflammatory cytokine upregulation and the infiltration of T cells, neutrophils, eosinophils, and macrophages into the lung. Overall, low-antigen-dose-induced ERD resembles FI-RSV-induced ERD, except that the former appears in the absence of detectable levels of viral replication and in the context of a Th1-biased immune response. Taken together, our observations reinforce the recent concept that vaccines developed for RSV-naïve individuals should be systematically tested under suboptimal dosing conditions.IMPORTANCE RSV poses a significant health care burden and is the leading cause of serious lower-respiratory-tract infections in young children. A formalin-inactivated RSV vaccine developed in the 1960s not only showed a complete lack of efficacy against RSV infection but also induced severe lung disease enhancement in vaccinated children. Since then, establishing safety in preclinical models has been one of the major challenges to RSV vaccine development. We recently observed in the cotton rat model that suboptimal immunizations with RSV fusion protein could induce lung disease enhancement. In the present study, we extended suboptimal dosing evaluation to the mouse model. We confirmed the induction of lung disease enhancement by vaccinations with low antigen doses and dissected the associated immune mechanisms. Our results stress the need to evaluate suboptimal dosing for any new RSV vaccine candidate developed for seronegative infants.
Collapse
Affiliation(s)
| | - Angie Snell Bennett
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | | | - Leigh Hostetler
- Laboratory Animal Resources, MedImmune, Gaithersburg, Maryland, USA
| | - Sean K Maynard
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | - Michelle Lazzaro
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | - Lily I Cheng
- Pathology Department, MedImmune, Gaithersburg, Maryland, USA
| | - Terrence O'Day
- Statistical Sciences, MedImmune, Gaithersburg, Maryland, USA
| | - Corinne Cayatte
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| |
Collapse
|
22
|
Kotomina T, Isakova-Sivak I, Matyushenko V, Kim KH, Lee Y, Jung YJ, Kang SM, Rudenko L. Recombinant live attenuated influenza vaccine viruses carrying CD8 T-cell epitopes of respiratory syncytial virus protect mice against both pathogens without inflammatory disease. Antiviral Res 2019; 168:9-17. [PMID: 31075351 PMCID: PMC6620116 DOI: 10.1016/j.antiviral.2019.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/29/2022]
Abstract
Respiratory syncytial virus (RSV) is the most common cause of lower respiratory disease in young children, elderly and immunocompromised adults. There is no licensed vaccine against RSV although development of an effective and safe RSV vaccine has been a high priority for several decades. Among the various vaccine platforms, the viral-vectored RSV vaccines based on licensed cold-adapted live attenuated influenza vaccine (LAIV) might offer an advantage of inducing adequate mucosal CD8 T cell immunity at the infection site of respiratory pathogens. We constructed two recombinant LAIV viruses expressing immunodominant T-cell epitopes of RSV M2-1 protein. The results in this study provide evidence that RSV CD8 T cell epitopes delivered by LAIV viral vector could confer protection against RSV infection without causing pulmonary eosinophilia and inflammatory RSV disease in mice. In addition, these chimeric LAIV-RSV vaccines retained their attenuated phenotype and ability to protect against virulent influenza virus, thus providing a unique approach to fight against two dangerous respiratory viral pathogens using a single vaccine preparation.
Collapse
Affiliation(s)
- Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia.
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, 12 Acad. Pavlov Street, 197376, Russia
| |
Collapse
|
23
|
Simon SCS, Utikal J, Umansky V. Opposing roles of eosinophils in cancer. Cancer Immunol Immunother 2019; 68:823-833. [PMID: 30302498 PMCID: PMC11028063 DOI: 10.1007/s00262-018-2255-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/28/2018] [Indexed: 12/13/2022]
Abstract
Eosinophils are a subset of granulocytes mostly known for their ability to combat parasites and induce allergy. Although they were described to be related to cancer more than 100 years ago, their role in tumors is still undefined. Recent observations revealed that they display regulatory functions towards other immune cell subsets in the tumor microenvironment or direct cytotoxic functions against tumor cells, leading to either antitumor or protumor effects. This paradoxical role of eosinophils was suggested to be dependent on the different factors in the TME. In addition, the clinical relevance of these cells has been recently addressed. In most cases, the accumulation of eosinophils both in the tumor tissue, called tumor-associated tissue eosinophilia, and in the peripheral blood were reported to be prognostic markers for a better outcome of cancer patients. In immunotherapy of cancer, particularly in therapy with immune checkpoint inhibitors, eosinophils were even shown to be a potential predictive marker for a beneficial clinical response. A better understanding of their role in cancer progression will help to establish them as prognostic and predictive markers and to design strategies for targeting eosinophils.
Collapse
Affiliation(s)
- Sonja C S Simon
- Skin Cancer Unit, Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, Clinical Cooperation Unit Dermato-Oncology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
24
|
Respiratory syncytial virus F and G protein core fragments fused to HBsAg-binding protein (SBP) induce a Th1-dominant immune response without vaccine-enhanced disease. Int Immunol 2018; 31:199-209. [DOI: 10.1093/intimm/dxy078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 11/16/2018] [Indexed: 12/28/2022] Open
|
25
|
Wacht G, Poirot A, Charles AL, Radosavljevic M, Uring-Lambert B, de Blay F, Geny B, Bahram S, Barnig C. FACS - based isolation of human eosinophils allows purification of high quality RNA. J Immunol Methods 2018; 463:47-53. [PMID: 30217720 DOI: 10.1016/j.jim.2018.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 08/18/2018] [Accepted: 09/06/2018] [Indexed: 12/25/2022]
Abstract
Here we extensively describe a FACS-based protocol for isolating intact non-stained human eosinophils from peripheral blood; a stop forward from our recently published initial study. This method of purification could be accomplished in <3 h with only small volumes of whole blood necessary, even in healthy subjects generally exhibiting low levels of circulating eosinophils. Eosinophil activation during the isolation steps appeared to be minimal and this purification procedure yielded high quality RNA. Moreover, these FACS-isolated eosinophils had prolonged viability in culture and were suitable for further activation assays.
Collapse
Affiliation(s)
- Guillaume Wacht
- Laboratoire Central d'Immunologie, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Nouvel Hôpital Civil, 67091 Strasbourg Cedex, France; EA 3072, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, 67085 Strasbourg Cedex, France
| | - Anh Poirot
- Pulmonology Unit, Department of Chest Disease, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University Hospital, 67091 Strasbourg Cedex, France
| | - Anne-Laure Charles
- EA 3072, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, 67085 Strasbourg Cedex, France
| | - Mirjana Radosavljevic
- Laboratoire Central d'Immunologie, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Nouvel Hôpital Civil, 67091 Strasbourg Cedex, France; INSERM UMR S_1109, ImmunoRhumatologie Moléculaire, Labex Transplantex, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, University de Strasbourg, 67085 Strasbourg, France
| | - Béatrice Uring-Lambert
- Laboratoire Central d'Immunologie, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Nouvel Hôpital Civil, 67091 Strasbourg Cedex, France
| | - Frédéric de Blay
- EA 3072, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, 67085 Strasbourg Cedex, France; Pulmonology Unit, Department of Chest Disease, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University Hospital, 67091 Strasbourg Cedex, France
| | - Bernard Geny
- EA 3072, Fédération de Médecine Translationnelle de Strasbourg, University of Strasbourg, 67085 Strasbourg Cedex, France; Physiology Unit, Department of Chest Disease, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University Hospital, 67091 Strasbourg Cedex, France
| | - Seiamak Bahram
- Laboratoire Central d'Immunologie, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Nouvel Hôpital Civil, 67091 Strasbourg Cedex, France; INSERM UMR S_1109, ImmunoRhumatologie Moléculaire, Labex Transplantex, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, University de Strasbourg, 67085 Strasbourg, France
| | - Cindy Barnig
- Laboratoire Central d'Immunologie, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Nouvel Hôpital Civil, 67091 Strasbourg Cedex, France; Pulmonology Unit, Department of Chest Disease, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, Strasbourg University Hospital, 67091 Strasbourg Cedex, France; INSERM UMR S_1109, ImmunoRhumatologie Moléculaire, Labex Transplantex, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg, University de Strasbourg, 67085 Strasbourg, France.
| |
Collapse
|
26
|
Miller MH, Shehat MG, Alcedo KP, Spinel LP, Soulakova J, Tigno-Aranjuez JT. Frontline Science: RIP2 promotes house dust mite-induced allergic airway inflammation. J Leukoc Biol 2018; 104:447-459. [PMID: 30052281 PMCID: PMC6113092 DOI: 10.1002/jlb.4hi0118-017rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 12/16/2022] Open
Abstract
House dust mites (HDMs) are one of the most significant environmental allergens in the establishment of the so-called "Atopic March." It is known that the immune response to HDM is Th2 dominant, but the innate mechanisms leading to HDM-induced type 2 responses are still not completely understood. A number of innate immune receptors have been implicated in the response to HDM including toll-like receptors, C-type lectin receptors, and protease activated receptors. NOD2 is a member of the NOD-like receptor family, which has been reported to be involved in the establishment of type 2 immunity and in blocking respiratory tolerance. NOD2 mediates its effects through its downstream effector kinase, receptor interacting protein (RIP2). It has not been shown if RIP2 is involved in the innate response to HDM and in the resulting generation of type 2 immunity. Furthermore, the role of RIP2 in modulating allergic airway inflammation has been controversial. In this study, we show that RIP2 is activated in airway epithelial cells in response to HDM and is important for the production of CCL2. Using a murine HDM asthma model, we demonstrate that lung pathology, local airway inflammation, inflammatory cytokines, HDM-specific IgG1 antibody production, and HDM-specific Th2 responses are all reduced in RIP2 knockout mice compared to WT animals. These data illustrate that RIP2 can be activated by a relevant allergic stimulus and that such activation can contribute to allergic airway inflammation. These findings also suggest that RIP2 inhibitors might have some efficacy in down-regulating the inflammatory response in type 2 dominated diseases.
Collapse
Affiliation(s)
- Madelyn H Miller
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Michael G Shehat
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Karel P Alcedo
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lina P Spinel
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Julia Soulakova
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| | - Justine T Tigno-Aranjuez
- Immunity and Pathogenesis Division, Burnett School of Biomedical Sciences, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
27
|
Skalski JH, Limon JJ, Sharma P, Gargus MD, Nguyen C, Tang J, Coelho AL, Hogaboam CM, Crother TR, Underhill DM. Expansion of commensal fungus Wallemia mellicola in the gastrointestinal mycobiota enhances the severity of allergic airway disease in mice. PLoS Pathog 2018; 14:e1007260. [PMID: 30235351 PMCID: PMC6147580 DOI: 10.1371/journal.ppat.1007260] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/06/2018] [Indexed: 12/26/2022] Open
Abstract
The gastrointestinal microbiota influences immune function throughout the body. The gut-lung axis refers to the concept that alterations of gut commensal microorganisms can have a distant effect on immune function in the lung. Overgrowth of intestinal Candida albicans has been previously observed to exacerbate allergic airways disease in mice, but whether subtler changes in intestinal fungal microbiota can affect allergic airways disease is less clear. In this study we have investigated the effects of the population expansion of commensal fungus Wallemia mellicola without overgrowth of the total fungal community. Wallemia spp. are commonly found as a minor component of the commensal gastrointestinal mycobiota in both humans and mice. Mice with an unaltered gut microbiota community resist population expansion when gavaged with W. mellicola; however, transient antibiotic depletion of gut microbiota creates a window of opportunity for expansion of W. mellicola following delivery of live spores to the gastrointestinal tract. This phenomenon is not universal as other commensal fungi (Aspergillus amstelodami, Epicoccum nigrum) do not expand when delivered to mice with antibiotic-depleted microbiota. Mice with Wallemia-expanded gut mycobiota experienced altered pulmonary immune responses to inhaled aeroallergens. Specifically, after induction of allergic airways disease with intratracheal house dust mite (HDM) antigen, mice demonstrated enhanced eosinophilic airway infiltration, airway hyperresponsiveness (AHR) to methacholine challenge, goblet cell hyperplasia, elevated bronchoalveolar lavage IL-5, and enhanced serum HDM IgG1. This phenomenon occurred with no detectable Wallemia in the lung. Targeted amplicon sequencing analysis of the gastrointestinal mycobiota revealed that expansion of W. mellicola in the gut was associated with additional alterations of bacterial and fungal commensal communities. We therefore colonized fungus-free Altered Schaedler Flora (ASF) mice with W. mellicola. ASF mice colonized with W. mellicola experienced enhanced severity of allergic airways disease compared to fungus-free control ASF mice without changes in bacterial community composition.
Collapse
Affiliation(s)
- Joseph H. Skalski
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, and the Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jose J. Limon
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, and the Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Purnima Sharma
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, and the Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Matthew D. Gargus
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, and the Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Christopher Nguyen
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, and the Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Jie Tang
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Ana Lucia Coelho
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Cory M. Hogaboam
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Timothy R. Crother
- Division of Pediatric Infectious Diseases, Department of Medicine, and the Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - David M. Underhill
- F. Widjaja Foundation Inflammatory Bowel & Immunobiology Research Institute, and the Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| |
Collapse
|
28
|
O’Sullivan JA, Wei Y, Carroll DJ, Moreno-Vinasco L, Cao Y, Zhang F, Lee JJ, Zhu Z, Bochner BS. Frontline Science: Characterization of a novel mouse strain expressing human Siglec-8 only on eosinophils. J Leukoc Biol 2018; 104:11-19. [PMID: 29601103 PMCID: PMC6013361 DOI: 10.1002/jlb.2hi0917-391r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 02/08/2018] [Accepted: 02/12/2018] [Indexed: 01/01/2023] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is a human cell surface protein expressed exclusively on eosinophils, mast cells, and basophils that, when engaged, induces eosinophil apoptosis and inhibits mast cell mediator release. This makes Siglec-8 a promising therapeutic target to treat diseases involving these cell types. However, preclinical studies of Siglec-8 targeting in vivo are lacking because this protein is only found in humans, apes, and some monkeys. Therefore, we have developed a mouse strain in which SIGLEC8 transcription is activated by Cre recombinase and have crossed this mouse with the eoCre mouse to achieve eosinophil-specific expression. We confirmed that Siglec-8 is expressed exclusively on the surface of mature eosinophils in multiple tissues at levels comparable to those on human blood eosinophils. Following ovalbumin sensitization and airway challenge, Siglec-8 knock-in mice generated a pattern of allergic lung inflammation indistinguishable from that of littermate controls, suggesting that Siglec-8 expression within the eosinophil compartment does not alter allergic eosinophilic inflammation. Using bone marrow from these mice, we demonstrated that, during maturation, Siglec-8 expression occurs well before the late eosinophil developmental marker C-C motif chemokine receptor 3, consistent with eoCre expression. Antibody ligation of the receptor induces Siglec-8 endocytosis and alters the phosphotyrosine profile of these cells, indicative of productive signaling. Finally, we demonstrated that mouse eosinophils expressing Siglec-8 undergo cell death when the receptor is engaged, further evidence that Siglec-8 is functional on these cells. These mice should prove useful to investigate Siglec-8 biology and targeting in vivo in a variety of eosinophilic disease models.
Collapse
Affiliation(s)
- Jeremy A. O’Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yadong Wei
- Section of Allergy and Clinical Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Daniela J. Carroll
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Liliana Moreno-Vinasco
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yun Cao
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Fengrui Zhang
- Section of Allergy and Clinical Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - James. J. Lee
- Division of Pulmonary Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Zhou Zhu
- Section of Allergy and Clinical Immunology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Bruce S. Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
29
|
Forkuo GS, Nieman AN, Kodali R, Zahn NM, Li G, Rashid Roni MS, Stephen MR, Harris TW, Jahan R, Guthrie ML, Yu OB, Fisher JL, Yocum GT, Emala CW, Steeber DA, Stafford DC, Cook JM, Arnold LA. A Novel Orally Available Asthma Drug Candidate That Reduces Smooth Muscle Constriction and Inflammation by Targeting GABA A Receptors in the Lung. Mol Pharm 2018; 15:1766-1777. [PMID: 29578347 PMCID: PMC5954213 DOI: 10.1021/acs.molpharmaceut.7b01013] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We describe lead compound MIDD0301 for the oral treatment of asthma based on previously developed positive allosteric α5β3γ2 selective GABAA receptor (GABAAR) ligands. MIDD0301 relaxed airway smooth muscle at single micromolar concentrations as demonstrated with ex vivo guinea pig tracheal rings. MIDD0301 also attenuated airway hyperresponsiveness (AHR) in an ovalbumin murine model of asthma by oral administration. Reduced numbers of eosinophils and macrophages were observed in mouse bronchoalveolar lavage fluid without changing mucous metaplasia. Importantly, lung cytokine expression of IL-17A, IL-4, and TNF-α were reduced for MIDD0301-treated mice without changing antiinflammatory cytokine IL-10 levels. Automated patch clamp confirmed amplification of GABA induced current mediated by α1-3,5β3γ2 GABAARs in the presence of MIDD0301. Pharmacodynamically, transmembrane currents of ex vivo CD4+ T cells from asthmatic mice were potentiated by MIDD0301 in the presence of GABA. The number of CD4+ T cells observed in the lung of MIDD0301-treated mice were reduced by an oral treatment of 20 mg/kg b.i.d. for 5 days. A half-life of almost 14 h was demonstrated by pharmacokinetic studies (PK) with no adverse CNS effects when treated mice were subjected to sensorimotor studies using the rotarod. PK studies also confirmed very low brain distribution. In conclusion, MIDD0301 represents a safe and improved oral asthma drug candidate that relaxes airway smooth muscle and attenuates inflammation in the lung leading to a reduction of AHR at a dosage lower than earlier reported GABAAR ligands.
Collapse
Affiliation(s)
- Gloria S. Forkuo
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Amanda N. Nieman
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Revathi Kodali
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Nicolas M. Zahn
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Guanguan Li
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - M. S. Rashid Roni
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Michael Rajesh Stephen
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Ted W. Harris
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Rajwana Jahan
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Margaret L. Guthrie
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Olivia B. Yu
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Janet L. Fisher
- Department of Pharmacology, Physiology & Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina 29208, United States
| | - Gene T. Yocum
- Department of Anesthesiology, Columbia University, New York, New York 10032, United States
| | - Charles W. Emala
- Department of Anesthesiology, Columbia University, New York, New York 10032, United States
| | - Douglas A. Steeber
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Douglas C. Stafford
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - James M. Cook
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| | - Leggy A. Arnold
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, Wisconsin 53211, United States
| |
Collapse
|
30
|
Yamaguchi M, Samuchiwal SK, Quehenberger O, Boyce JA, Balestrieri B. Macrophages regulate lung ILC2 activation via Pla2g5-dependent mechanisms. Mucosal Immunol 2018; 11:615-626. [PMID: 29346348 PMCID: PMC5976507 DOI: 10.1038/mi.2017.99] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/17/2017] [Indexed: 02/04/2023]
Abstract
Group V phospholipase A2 (Pla2g5) is a lipid-generating enzyme necessary for macrophage effector functions in pulmonary inflammation. However, the lipid mediators involved and their cellular targets have not been identified. Mice lacking Pla2g5 showed markedly reduced lung ILC2 activation and eosinophilia following repetitive Alternaria Alternata inhalation. While Pla2g5-null mice had Wt levels of immediate IL-33 release after one Alternaria dose, they failed to upregulate IL-33 in macrophages following repeated Alternaria administration. Unexpectedly, while adoptive transfer of bone marrow-derived (BM)-macrophages restored ILC2 activation and eosinophilia in Alternaria-exposed Pla2g5-null mice, exogenous IL-33 did not. Conversely, transfers of Pla2g5-null BM-macrophages reduced inflammation in Alternaria-exposed Wt mice. Mass spectrometry analysis of free fatty acids (FFAs) demonstrated significantly reduced FFAs (including linoleic acid (LA) and oleic acid (OA)) in lung and BM-macrophages lacking Pla2g5. Exogenous administration of LA or LA+OA to Wt mice sharply potentiated IL-33-induced lung eosinophilia and ILC2 expansion in vitro and in vivo. In contrast, OA potentiated IL-33-induced inflammation and ILC2 expansion in Pla2g5-null mice, but LA was inactive both in vivo and in vitro. Notably, Pla2g5-null ILC2s showed significantly reduced expression of the FFA-receptor-1 compared to Wt ILC2s. Thus, macrophage-associated Pla2g5 contributes significantly to type-2 immunity through regulation of IL-33 induction and FFA-driven ILC2 activation.
Collapse
Affiliation(s)
- Munehiro Yamaguchi
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Sachin K. Samuchiwal
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Oswald Quehenberger
- Department of Medicine, Department of Pharmacology, University of California, San Diego, CA 92093, USA
| | - Joshua A. Boyce
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Barbara Balestrieri
- Department of Medicine, Harvard Medical School, Jeff and Penny Vinik Center for Allergic Disease Research, Division of Rheumatology, Immunology and Allergy, Brigham and Women’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
31
|
Matsuda M, Tabuchi Y, Nishimura K, Nakamura Y, Sekioka T, Kadode M, Kawabata K, Nabe T. Increased expression of CysLT 2 receptors in the lung of asthmatic mice and role in allergic responses. Prostaglandins Leukot Essent Fatty Acids 2018; 131:24-31. [PMID: 29628047 DOI: 10.1016/j.plefa.2018.03.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/13/2018] [Accepted: 03/21/2018] [Indexed: 12/23/2022]
Abstract
Compared with CysLT1 receptors, the functional role of CysLT2 receptors in asthma has not been clarified. The purpose of this study was to determine 1) whether CysLT2 receptors are expressed in the lung of mice and if expression increases in asthmatic mice, and 2) whether CysLT2 receptors are involved in allergic leukocyte infiltration into the lung and in the development of airway remodeling in asthmatic mice. BALB/c mice were sensitized with ovalbumin (OVA) + Al(OH)3, and intratracheally challenged with OVA 4 times. Lung tissue was isolated before and after the 4th OVA challenge for detection of CysLT2 receptors by immunohistochemistry and flow cytometry. The effect of a CysLT2 receptor antagonist BayCysLT2RA on multiple antigen challenge-induced leukocyte infiltration into the lung and the development of airway remodeling was evaluated. Even in non-challenged mice, CysLT2 receptors were expressed in bronchial smooth muscle. After multiple challenges, expression was also observed in leukocytes infiltrating into alveolar spaces. CysLT2R+ leukocytes included alveolar macrophages, conventional dendritic cells, and eosinophils. BayCysLT2RA significantly inhibited multiple antigen challenge-induced increases in eosinophils and mononuclear cells in the lung. The development of airway remodeling was tended to be suppressed by CysLT2 receptor antagonist. In conclusion, CysLT2 receptors were constitutively expressed in the lung, and expression was strengthened in asthmatic mice. Activation of CysLT2 receptors was functionally involved in allergic leukocyte infiltration into the lung. The CysLT2 receptor can be a molecular target for the development of new pharmacotherapies for asthma.
Collapse
Affiliation(s)
- Masaya Matsuda
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Yuki Tabuchi
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Kazuma Nishimura
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Yuri Nakamura
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan
| | - Tomohiko Sekioka
- Minase Research Institute, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Michiaki Kadode
- Minase Research Institute, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Kazuhito Kawabata
- Minase Research Institute, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Takeshi Nabe
- Department of Immunopharmacology, Faculty of Pharmaceutical Sciences, Setsunan University, Osaka, Japan.
| |
Collapse
|
32
|
Geslewitz WE, Percopo CM, Rosenberg HF. FACS isolation of live mouse eosinophils at high purity via a protocol that does not target Siglec F. J Immunol Methods 2017; 454:27-31. [PMID: 29253503 DOI: 10.1016/j.jim.2017.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 11/21/2017] [Accepted: 12/07/2017] [Indexed: 01/01/2023]
Abstract
Flow cytometry protocols designed to identify mouse eosinophils typically target Siglec F, an α-2,3-sialic acid binding transmembrane protein expressed universally on cells of this lineage. While a convenient target, antibody-mediated ligation of Siglec F induces eosinophil apoptosis, which limits its usefulness for isolations that are to be followed by functional and/or gene expression studies. We present here a method for FACS isolation which does not target Siglec F and likewise utilizes no antibodies targeting IL5Rα (CD125) or CCR3. Single cell suspensions are prepared from lungs of mice that were sensitized and challenged with Aspergillus fumigatus antigens; eosinophils were identified and isolated by FACS as live SSChi/FSChi CD11c-Gr1-/loMHCII- cells. This strategy was also effective for eosinophil isolation from the lungs of IL5tg mice. Purity by visual inspection of stained cytospin preparations and by Siglec F-diagnostic flow cytometry was 98-99% and 97-99%, respectively. Eosinophils isolated by this method (yield, ~4×106/mouse) generated high-quality RNA suitable for gene expression analysis.
Collapse
Affiliation(s)
- Wendy E Geslewitz
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Caroline M Percopo
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Helene F Rosenberg
- Inflammation Immunobiology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
33
|
Abstract
Eosinophils are a prominent cell type in particular host responses such as the response to helminth infection and allergic disease. Their effector functions have been attributed to their capacity to release cationic proteins stored in cytoplasmic granules by degranulation. However, eosinophils are now being recognized for more varied functions in previously underappreciated diverse tissue sites, based on the ability of eosinophils to release cytokines (often preformed) that mediate a broad range of activities into the local environment. In this Review, we consider evolving insights into the tissue distribution of eosinophils and their functional immunobiology, which enable eosinophils to secrete in a selective manner cytokines and other mediators that have diverse, 'non-effector' functions in health and disease.
Collapse
Affiliation(s)
- Peter F Weller
- Division of Allergy and Inflammation, Harvard Medical School, Beth Israel Deaconess Medical Center, CLS 943, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| | - Lisa A Spencer
- Division of Allergy and Inflammation, Harvard Medical School, Beth Israel Deaconess Medical Center, CLS 943, 330 Brookline Avenue, Boston, Massachusetts 02215, USA
| |
Collapse
|
34
|
Alteration of Inflammatory Mediators in the Upper and Lower Airways under Chronic Intermittent Hypoxia: Preliminary Animal Study. Mediators Inflamm 2017; 2017:4327237. [PMID: 29038619 PMCID: PMC5606044 DOI: 10.1155/2017/4327237] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/20/2017] [Accepted: 08/02/2017] [Indexed: 11/17/2022] Open
Abstract
Purpose We hypothesized that CIH may affect the upper airway immune system and aimed to verify whether CIH can induce airway inflammation in a murine obstructive sleep apnea (OSA) model. Methods C57BL6 male mice were exposed to intermittent hypoxia (CIH group; 5 ~ 21% FiO2, 120 sec cycles, 12 h/d, n = 6) or room air (Sham group, n = 6) for up to 4 weeks in identical chambers. Nasal and lung tissues and lavage fluid were collected and analyzed by multiplex assay. Lung lavage fluid was also utilized for FACS analysis to determine eosinophil count. Results We determined the protein level of 24 different cytokines, chemokines, and inflammatory mediators. Among various cytokines, levels of IL-1α, IL-1β, IL-4, IL-6, and IL-13 were significantly elevated in nose or lung tissue from the CIH group. In addition, MCP-1 and periostin were elevated in nose and lung tissue and lavage fluid from the CIH group. Conclusions CIH for 4 weeks altered the levels of inflammatory mediators in both the nose and lungs of mouse model. We suggest that the airway immune system may be deteriorated by CIH and allergic inflammation in the upper or lower airway could be worsened by sleep apnea.
Collapse
|
35
|
Wang X, Wu W, Zhang W, Leland Booth J, Duggan ES, Tian L, More S, Zhao YD, Sawh RN, Liu L, Zou MH, Metcalf JP. RIG-I overexpression decreases mortality of cigarette smoke exposed mice during influenza A virus infection. Respir Res 2017; 18:166. [PMID: 28865477 PMCID: PMC5581920 DOI: 10.1186/s12931-017-0649-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 08/25/2017] [Indexed: 12/22/2022] Open
Abstract
Background Retinoic acid-inducible gene I (RIG-I) is an important regulator of virus-induced antiviral interferons (IFNs) and proinflammatory cytokines which participate in clearing viral infections. Cigarette smoke (CS) exposure increases the frequency and severity of respiratory tract infections. Methods We generated a RIG-I transgenic (TG) mouse strain that expresses the RIG-I gene product under the control of the human lung specific surfactant protein C promoter. We compared the mortality and host immune responses of RIG-I TG mice and their litter-matched wild type (WT) mice following challenge with influenza A virus (IAV). Results RIG-I overexpression increased survival of IAV-infected mice. CS exposure increased mortality in WT mice infected with IAV. Remarkably, the effect of RIG-I overexpression on survival during IAV infection was enhanced in CS-exposed animals. CS-exposed IAV-infected WT mice had a suppressed innate response profile in the lung compared to sham-exposed IAV-infected WT mice in terms of the protein concentration, total cell count and inflammatory cell composition in the bronchoalveolar lavage fluid. RIG-I overexpression restored the innate immune response in CS-exposed mice to that seen in sham-exposed WT mice during IAV infection, and is likely responsible for enhanced survival in RIG-I TG mice as restoration preceded death of the animals. Conclusions Our results demonstrate that RIG-I overexpression in mice is protective for CS enhanced susceptibility of smokers to influenza infection, and that CS mediated RIG-I suppression may be partially responsible for the increased morbidity and mortality of the mice exposed to IAV. Thus, optimizing the RIG-I response may be an important treatment strategy for CS-enhanced lung infections, particularly those due to IAV.
Collapse
Affiliation(s)
- Xiaoqiu Wang
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Wenxin Wu
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Wei Zhang
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - J Leland Booth
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Elizabeth S Duggan
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Lili Tian
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sunil More
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Yan D Zhao
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ravindranauth N Sawh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Lin Liu
- The Lundberg-Kienlen Lung Biology and Toxicology Laboratory, Department of Physiological Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK, USA
| | - Ming-Hui Zou
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, GA, USA
| | - Jordan P Metcalf
- Pulmonary and Critical Care Division, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA. .,Veterans Affairs Medical Center, Oklahoma City, OK, USA. .,Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
36
|
Fa Z, Xie Q, Fang W, Zhang H, Zhang H, Xu J, Pan W, Xu J, Olszewski MA, Deng X, Liao W. RIPK3/Fas-Associated Death Domain Axis Regulates Pulmonary Immunopathology to Cryptococcal Infection Independent of Necroptosis. Front Immunol 2017; 8:1055. [PMID: 28919893 PMCID: PMC5585137 DOI: 10.3389/fimmu.2017.01055] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 08/14/2017] [Indexed: 01/08/2023] Open
Abstract
Fas-associated death domain (FADD) and receptor interacting protein kinase 3 (RIPK3) are multifunctional regulators of cell death and immune response. Using a mouse model of cryptococcal infection, the roles of FADD and RIPK3 in anti-cryptococcal defense were investigated. Deletion of RIPK3 alone led to increased inflammatory cytokine production in the Cryptococcus neoformans-infected lungs, but in combination with FADD deletion, it led to a robust Th1-biased response with M1-biased macrophage activation. Rather than being protective, these responses led to paradoxical C. neoformans expansion and rapid clinical deterioration in Ripk3−/− and Ripk3−/−Fadd−/− mice. The increased mortality of Ripk3−/− and even more accelerated mortality in Ripk3−/−Fadd−/− mice was attributed to profound pulmonary damage due to neutrophil-dominant infiltration with prominent upregulation of pro-inflammatory cytokines. This phenomenon was partially associated with selective alterations in the apoptotic frequency of some leukocyte subsets, such as eosinophils and neutrophils, in infected Ripk3−/−Fadd−/− mice. In conclusion, our study shows that RIPK3 in concert with FADD serve as physiological “brakes,” preventing the development of excessive inflammation and Th1 bias, which in turn contributes to pulmonary damage and defective fungal clearance. This novel link between the protective effect of FADD and RIPK3 in antifungal defense and sustenance of immune homeostasis may be important for the development of novel immunomodulatory therapies against invasive fungal infections.
Collapse
Affiliation(s)
- Zhenzong Fa
- PLA Key Laboratory of Mycosis, Department of Dermatology and Venereology, Changzheng Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Institute of Medical Mycology, Second Military Medical University, Shanghai, China.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, United States
| | - Qun Xie
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China.,Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei Fang
- Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Institute of Medical Mycology, Second Military Medical University, Shanghai, China.,Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haibing Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Haiwei Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jintao Xu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, United States
| | - Weihua Pan
- PLA Key Laboratory of Mycosis, Department of Dermatology and Venereology, Changzheng Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Institute of Medical Mycology, Second Military Medical University, Shanghai, China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Michal A Olszewski
- Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Institute of Medical Mycology, Second Military Medical University, Shanghai, China.,Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, United States
| | - Xiaoming Deng
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wanqing Liao
- PLA Key Laboratory of Mycosis, Department of Dermatology and Venereology, Changzheng Hospital, Shanghai, China.,Shanghai Key Laboratory of Molecular Medical Mycology, Shanghai Institute of Medical Mycology, Second Military Medical University, Shanghai, China
| |
Collapse
|
37
|
Corthésy B, Bioley G. Therapeutic intranasal instillation of allergen-loaded microbubbles suppresses experimental allergic asthma in mice. Biomaterials 2017; 142:41-51. [PMID: 28727997 DOI: 10.1016/j.biomaterials.2017.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/20/2017] [Accepted: 07/09/2017] [Indexed: 12/18/2022]
Abstract
Despite proven efficiency, subcutaneous immunotherapy for aeroallergens is impaired by the duration of the protocol, the repeated injections and potential side-effects associated with the doses of allergen administered. Intranasal delivery of immunotherapeutic agents may overcome several of these drawbacks, provided that an efficient allergen delivery vehicle can be identified. This study evaluates whether intranasally delivered gas-filled microbubble (MB)-associated ovalbumin (OVA), used as a model allergen, can serve as a therapeutic treatment in a mouse model of established allergic asthma. Lung and systemic production of pro-tolerogenic markers, including Foxp3+ CD4 T cells, IL-10, and TGF-β, as well as the Th1-type cytokine IFN-γ, was observed after intranasal immunization with OVA-MB. Post-treatment, aerosol-sensitized mice exhibited the same pattern of markers. Moreover, decrease of eosinophils and neutrophils in BALs, lower frequencies of Th2 cytokine- and IL-17-producing CD4 T cells in lungs and reduced specific IgE in BALs and sera after allergen challenge were observed. Concomitantly, lung resistance and mucus production diminished in OVA-MB-treated animals. Thus, therapeutic intranasal administration of OVA-MBs in established experimental allergic asthma allows modulating pathology-associated immune and physiological parameters usually triggered after exposure to the allergen.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, University State Hospital (CHUV), Epalinges, Switzerland
| | - Gilles Bioley
- R&D Laboratory, Division of Immunology and Allergy, University State Hospital (CHUV), Epalinges, Switzerland.
| |
Collapse
|
38
|
Diny NL, Rose NR, Čiháková D. Eosinophils in Autoimmune Diseases. Front Immunol 2017; 8:484. [PMID: 28496445 PMCID: PMC5406413 DOI: 10.3389/fimmu.2017.00484] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/07/2017] [Indexed: 12/15/2022] Open
Abstract
Eosinophils are multifunctional granulocytes that contribute to initiation and modulation of inflammation. Their role in asthma and parasitic infections has long been recognized. Growing evidence now reveals a role for eosinophils in autoimmune diseases. In this review, we summarize the function of eosinophils in inflammatory bowel diseases, neuromyelitis optica, bullous pemphigoid, autoimmune myocarditis, primary biliary cirrhosis, eosinophilic granulomatosis with polyangiitis, and other autoimmune diseases. Clinical studies, eosinophil-targeted therapies, and experimental models have contributed to our understanding of the regulation and function of eosinophils in these diseases. By examining the role of eosinophils in autoimmune diseases of different organs, we can identify common pathogenic mechanisms. These include degranulation of cytotoxic granule proteins, induction of antibody-dependent cell-mediated cytotoxicity, release of proteases degrading extracellular matrix, immune modulation through cytokines, antigen presentation, and prothrombotic functions. The association of eosinophilic diseases with autoimmune diseases is also examined, showing a possible increase in autoimmune diseases in patients with eosinophilic esophagitis, hypereosinophilic syndrome, and non-allergic asthma. Finally, we summarize key future research needs.
Collapse
Affiliation(s)
- Nicola L Diny
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Noel R Rose
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
39
|
Guerra ES, Lee CK, Specht CA, Yadav B, Huang H, Akalin A, Huh JR, Mueller C, Levitz SM. Central Role of IL-23 and IL-17 Producing Eosinophils as Immunomodulatory Effector Cells in Acute Pulmonary Aspergillosis and Allergic Asthma. PLoS Pathog 2017; 13:e1006175. [PMID: 28095479 PMCID: PMC5271415 DOI: 10.1371/journal.ppat.1006175] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 01/27/2017] [Accepted: 01/09/2017] [Indexed: 02/06/2023] Open
Abstract
Aspergillus fumigatus causes invasive pulmonary disease in immunocompromised hosts and allergic asthma in atopic individuals. We studied the contribution of lung eosinophils to these fungal diseases. By in vivo intracellular cytokine staining and confocal microscopy, we observed that eosinophils act as local sources of IL-23 and IL-17. Remarkably, mice lacking eosinophils had a >95% reduction in the percentage of lung IL-23p19+ cells as well as markedly reduced IL-23 heterodimer in lung lavage fluid. Eosinophils killed A. fumigatus conidia in vivo. Eosinopenic mice had higher mortality rates, decreased recruitment of inflammatory monocytes, and decreased expansion of lung macrophages after challenge with conidia. All of these functions underscore a potential protective role for eosinophils in acute aspergillosis. Given the postulated role for IL-17 in asthma pathogenesis, we assessed whether eosinophils could act as sources of IL-23 and IL-17 in models where mice were sensitized to either A. fumigatus antigens or ovalbumin (OVA). We found IL-23p19+ IL-17AF+ eosinophils in both allergic models. Moreover, close to 95% of IL-23p19+ cells and >90% of IL-17AF+ cells were identified as eosinophils. These data establish a new paradigm in acute and allergic aspergillosis whereby eosinophils act not only as effector cells but also as immunomodulatory cells driving the IL-23/IL-17 axis and contributing to inflammatory cell recruitment. The opportunistic fungus, Aspergillus fumigatus, causes a spectrum of diseases ranging from invasive aspergillosis in the severely immunosuppressed to allergic asthma in atopic individuals. Here we explored the contribution of eosinophils, a type of white blood cell, to host defenses and pathogenesis in murine models of invasive pulmonary aspergillosis and asthma. We found eosinophils co-produce the cytokines IL-23 and IL-17 in both aspergillosis models as well as a model of OVA-induced asthma. Eosinophils killed the conidia (spores) of A. fumigatus in vivo and mice that lacked eosinophils were more susceptible to invasive aspergillosis. These observations suggest eosinophils play a more prominent role in defenses against invasive pulmonary aspergillosis than heretofore appreciated and identify eosinophil-derived IL-23 and IL-17 as potential therapeutic targets in allergic asthma.
Collapse
Affiliation(s)
- Evelyn Santos Guerra
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Chrono K Lee
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Charles A Specht
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Bhawna Yadav
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Haibin Huang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Ali Akalin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Jun R Huh
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Christian Mueller
- Horae Gene Therapy Center and Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Stuart M Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, United States of America
| |
Collapse
|
40
|
Regulation of airway inflammation by Siglec-8 and Siglec-9 sialoglycan ligand expression. Curr Opin Allergy Clin Immunol 2016; 16:24-30. [PMID: 26694037 DOI: 10.1097/aci.0000000000000234] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Inflammatory cells involved in the allergic response, including eosinophils, mast cells, basophils, and neutrophils, express sialoglycan-binding proteins such as Siglec-8 and Siglec-9, which inhibit cell function and survival. The purpose of this review is to briefly discuss the biology of these siglecs and their ligands and consider their potential impact in pathology and treatment of chronic rhinosinusitis (CRS). RECENT FINDINGS Recent studies demonstrate the presence of ligands for Siglec-8 and Siglec-9 in sinonasal tissue from patients with CRS as well as healthy patients, suggesting that the immunoregulatory functions of siglecs may be triggered in sinus tissue in health and disease. SUMMARY Ligands for Siglec-8 and Siglec-9 may regulate the function of eosinophils, mast cells, neutrophils, and other cells in sinus mucosa. Therapeutic strategies that activate the anti-inflammatory effects of siglecs may dampen inflammation and disease in CRS patients.
Collapse
|
41
|
de Graaf CA, Choi J, Baldwin TM, Bolden JE, Fairfax KA, Robinson AJ, Biben C, Morgan C, Ramsay K, Ng AP, Kauppi M, Kruse EA, Sargeant TJ, Seidenman N, D'Amico A, D'Ombrain MC, Lucas EC, Koernig S, Baz Morelli A, Wilson MJ, Dower SK, Williams B, Heazlewood SY, Hu Y, Nilsson SK, Wu L, Smyth GK, Alexander WS, Hilton DJ. Haemopedia: An Expression Atlas of Murine Hematopoietic Cells. Stem Cell Reports 2016; 7:571-582. [PMID: 27499199 PMCID: PMC5031953 DOI: 10.1016/j.stemcr.2016.07.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/09/2016] [Accepted: 07/10/2016] [Indexed: 12/12/2022] Open
Abstract
Hematopoiesis is a multistage process involving the differentiation of stem and progenitor cells into distinct mature cell lineages. Here we present Haemopedia, an atlas of murine gene-expression data containing 54 hematopoietic cell types, covering all the mature lineages in hematopoiesis. We include rare cell populations such as eosinophils, mast cells, basophils, and megakaryocytes, and a broad collection of progenitor and stem cells. We show that lineage branching and maturation during hematopoiesis can be reconstructed using the expression patterns of small sets of genes. We also have identified genes with enriched expression in each of the mature blood cell lineages, many of which show conserved lineage-enriched expression in human hematopoiesis. We have created an online web portal called Haemosphere to make analyses of Haemopedia and other blood cell transcriptional datasets easier. This resource provides simple tools to interrogate gene-expression-based relationships between hematopoietic cell types and genes of interest.
Collapse
Affiliation(s)
- Carolyn A de Graaf
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Jarny Choi
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Tracey M Baldwin
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Jessica E Bolden
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kirsten A Fairfax
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Aaron J Robinson
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Christine Biben
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Clare Morgan
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kerry Ramsay
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Ashley P Ng
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Maria Kauppi
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Elizabeth A Kruse
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Tobias J Sargeant
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Nick Seidenman
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Angela D'Amico
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Marthe C D'Ombrain
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; CSL Limited, Parkville, VIC 3052, Australia
| | - Erin C Lucas
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | | | | | | | | | - Brenda Williams
- Biomedical Manufacturing, CSIRO Manufacturing, Clayton, VIC 3169, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Shen Y Heazlewood
- Biomedical Manufacturing, CSIRO Manufacturing, Clayton, VIC 3169, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Yifang Hu
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3010, Australia
| | - Susan K Nilsson
- Biomedical Manufacturing, CSIRO Manufacturing, Clayton, VIC 3169, Australia; Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Li Wu
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Tsinghua University School of Medicine, Beijing 100084, China
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3010, Australia; Department of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Warren S Alexander
- Cancer and Haematology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Douglas J Hilton
- Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
42
|
Mesnil C, Raulier S, Paulissen G, Xiao X, Birrell MA, Pirottin D, Janss T, Starkl P, Ramery E, Henket M, Schleich FN, Radermecker M, Thielemans K, Gillet L, Thiry M, Belvisi MG, Louis R, Desmet C, Marichal T, Bureau F. Lung-resident eosinophils represent a distinct regulatory eosinophil subset. J Clin Invest 2016; 126:3279-95. [PMID: 27548519 DOI: 10.1172/jci85664] [Citation(s) in RCA: 404] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 06/09/2016] [Indexed: 12/21/2022] Open
Abstract
Increases in eosinophil numbers are associated with infection and allergic diseases, including asthma, but there is also evidence that eosinophils contribute to homeostatic immune processes. In mice, the normal lung contains resident eosinophils (rEos), but their function has not been characterized. Here, we have reported that steady-state pulmonary rEos are IL-5-independent parenchymal Siglec-FintCD62L+CD101lo cells with a ring-shaped nucleus. During house dust mite-induced airway allergy, rEos features remained unchanged, and rEos were accompanied by recruited inflammatory eosinophils (iEos), which were defined as IL-5-dependent peribronchial Siglec-FhiCD62L-CD101hi cells with a segmented nucleus. Gene expression analyses revealed a more regulatory profile for rEos than for iEos, and correspondingly, mice lacking lung rEos showed an increase in Th2 cell responses to inhaled allergens. Such elevation of Th2 responses was linked to the ability of rEos, but not iEos, to inhibit the maturation, and therefore the pro-Th2 function, of allergen-loaded DCs. Finally, we determined that the parenchymal rEos found in nonasthmatic human lungs (Siglec-8+CD62L+IL-3Rlo cells) were phenotypically distinct from the iEos isolated from the sputa of eosinophilic asthmatic patients (Siglec-8+CD62LloIL-3Rhi cells), suggesting that our findings in mice are relevant to humans. In conclusion, our data define lung rEos as a distinct eosinophil subset with key homeostatic functions.
Collapse
|
43
|
Percopo CM, Brenner TA, Ma M, Kraemer LS, Hakeem RMA, Lee JJ, Rosenberg HF. SiglecF+Gr1hi eosinophils are a distinct subpopulation within the lungs of allergen-challenged mice. J Leukoc Biol 2016; 101:321-328. [PMID: 27531929 DOI: 10.1189/jlb.3a0416-166r] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 07/07/2016] [Accepted: 07/25/2016] [Indexed: 01/06/2023] Open
Abstract
Although eosinophils as a group are readily identified by their unique morphology and staining properties, flow cytometry provides an important means for identification of subgroups based on differential expression of distinct surface Ags. Here, we characterize an eosinophil subpopulation defined by high levels of expression of the neutrophil Ag Gr1 (CD45+CD11c-SiglecF+Gr1hi). SiglecF+Gr1hi eosinophils, distinct from the canonical SiglecF+Gr1- eosinophil population, were detected in allergen-challenged wild-type and granule protein-deficient (EPX-/- and MBP-1-/-) mice, but not in the eosinophil-deficient ΔdblGATA strain. In contrast to Gr1+ neutrophils, which express both cross-reacting Ags Ly6C and Ly6G, SiglecF+Gr1hi eosinophils from allergen-challenged lung tissue are uniquely Ly6G+ Although indistinguishable from the more-numerous SiglecF+Gr1- eosinophils under light microscopy, FACS-isolated populations revealed prominent differences in cytokine contents. The lymphocyte-targeting cytokines CXCL13 and IL-27 were identified only in the SiglecF+Gr1hi eosinophil population (at 3.9 and 4.8 pg/106 cells, respectively), as was the prominent proinflammatory mediator IL-13 (72 pg/106 cells). Interestingly, bone marrow-derived (SiglecF+), cultured eosinophils include a more substantial Gr1+ subpopulation (∼50%); Gr1+ bmEos includes primarily a single Ly6C+ and a smaller, double-positive (Ly6C+Ly6G+) population. Taken together, our findings characterize a distinct SiglecF+Gr1hi eosinophil subset in lungs of allergen-challenged, wild-type and granule protein-deficient mice. SiglecF+Gr1hi eosinophils from wild-type mice maintain a distinct subset of cytokines, including those active on B and T lymphocytes. These cytokines may facilitate eosinophil-mediated immunomodulatory responses in the allergen-challenged lung as well as in other distinct microenvironments.
Collapse
Affiliation(s)
- Caroline M Percopo
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Todd A Brenner
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michelle Ma
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Laura S Kraemer
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Reem M A Hakeem
- Molecular Signal Transduction Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA; and
| | | | - Helene F Rosenberg
- Inflammation Immunobiology Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
44
|
Zeidler-Erdely PC, Antonini JM, Meighan TG, Young SH, Eye TJ, Hammer MA, Erdely A. Comparison of cell counting methods in rodent pulmonary toxicity studies: automated and manual protocols and considerations for experimental design. Inhal Toxicol 2016; 28:410-20. [PMID: 27251196 DOI: 10.1080/08958378.2016.1189985] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Pulmonary toxicity studies often use bronchoalveolar lavage (BAL) to investigate potential adverse lung responses to a particulate exposure. The BAL cellular fraction is counted, using automated (i.e. Coulter Counter®), flow cytometry or manual (i.e. hemocytometer) methods, to determine inflammatory cell influx. The goal of the study was to compare the different counting methods to determine which is optimal for examining BAL cell influx after exposure by inhalation or intratracheal instillation (ITI) to different particles with varying inherent pulmonary toxicities in both rat and mouse models. General findings indicate that total BAL cell counts using the automated and manual methods tended to agree after inhalation or ITI exposure to particle samples that are relatively nontoxic or at later time points after exposure to a pneumotoxic particle when the response resolves. However, when the initial lung inflammation and cytotoxicity was high after exposure to a pneumotoxic particle, significant differences were observed when comparing cell counts from the automated, flow cytometry and manual methods. When using total BAL cell count for differential calculations from the automated method, depending on the cell diameter size range cutoff, the data suggest that the number of lung polymorphonuclear leukocytes (PMN) varies. Importantly, the automated counts, regardless of the size cutoff, still indicated a greater number of total lung PMN when compared with the manual method, which agreed more closely with flow cytometry. The results suggest that either the manual method or flow cytometry would be better suited for BAL studies where cytotoxicity is an unknown variable.
Collapse
Affiliation(s)
- Patti C Zeidler-Erdely
- a Health Effects Laboratory Division, National Institute for Occupational Safety and Health , Morgantown , WV , USA and
| | - James M Antonini
- a Health Effects Laboratory Division, National Institute for Occupational Safety and Health , Morgantown , WV , USA and
| | - Terence G Meighan
- a Health Effects Laboratory Division, National Institute for Occupational Safety and Health , Morgantown , WV , USA and
| | - Shih-Houng Young
- b Army Public Health Center (Provisional) , Aberdeen Proving Ground , MD , USA
| | - Tracy J Eye
- a Health Effects Laboratory Division, National Institute for Occupational Safety and Health , Morgantown , WV , USA and
| | - Mary Ann Hammer
- a Health Effects Laboratory Division, National Institute for Occupational Safety and Health , Morgantown , WV , USA and
| | - Aaron Erdely
- a Health Effects Laboratory Division, National Institute for Occupational Safety and Health , Morgantown , WV , USA and
| |
Collapse
|
45
|
Nayak AP, Green BJ, Lemons AR, Marshall NB, Goldsmith WT, Kashon ML, Anderson SE, Germolec DR, Beezhold DH. Subchronic exposures to fungal bioaerosols promotes allergic pulmonary inflammation in naïve mice. Clin Exp Allergy 2016; 46:861-70. [PMID: 26892490 DOI: 10.1111/cea.12724] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 02/05/2016] [Accepted: 02/07/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Epidemiological surveys indicate that occupants of mold contaminated environments are at increased risk of respiratory symptoms. The immunological mechanisms associated with these responses require further characterization. OBJECTIVE The aim of this study was to characterize the immunotoxicological outcomes following repeated inhalation of dry Aspergillus fumigatus spores aerosolized at concentrations potentially encountered in contaminated indoor environments. METHODS Aspergillus fumigatus spores were delivered to the lungs of naïve BALB/cJ mice housed in a multi-animal nose-only chamber twice a week for a period of 13 weeks. Mice were evaluated at 24 and 48 h post-exposure for histopathological changes in lung architecture, recruitment of specific immune cells to the airways, and serum antibody responses. RESULT Germinating A. fumigatus spores were observed in lungs along with persistent fungal debris in the perivascular regions of the lungs. Repeated exposures promoted pleocellular infiltration with concomitant epithelial mucus hypersecretion, goblet cell metaplasia, subepithelial fibrosis and enhanced airway hyperreactivity. Cellular infiltration in airways was predominated by CD4(+) T cells expressing the pro-allergic cytokine IL-13. Furthermore, our studies show that antifungal T cell responses (IFN-γ(+) or IL-17A(+) ) co-expressed IL-13, revealing a novel mechanism for the dysregulated immune response to inhaled fungi. Total IgE production was augmented in animals repeatedly exposed to A. fumigatus. CONCLUSIONS & CLINICAL RELEVANCE Repeated inhalation of fungal aerosols resulted in significant pulmonary pathology mediated by dynamic shifts in specific immune populations and their cytokines. These studies provide novel insights into the immunological mechanisms and targets that govern the health outcomes that result from repeated inhalation of fungal bioaerosols in contaminated environments.
Collapse
Affiliation(s)
- A P Nayak
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - B J Green
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - A R Lemons
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - N B Marshall
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - W T Goldsmith
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - M L Kashon
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - S E Anderson
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| | - D R Germolec
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - D H Beezhold
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, USA
| |
Collapse
|
46
|
Liu Y, Huang X, Yi X, He Y, Mylonakis E, Xi L. Detection of Talaromyces marneffei from Fresh Tissue of an Inhalational Murine Pulmonary Model Using Nested PCR. PLoS One 2016; 11:e0149634. [PMID: 26886887 PMCID: PMC4757548 DOI: 10.1371/journal.pone.0149634] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/03/2016] [Indexed: 11/18/2022] Open
Abstract
Penicilliosis marneffei, often consecutive to the aspiration of Talaromyces marneffei (Penicillium marneffei), continues to be one of the significant causes of morbidity and mortality in immunocompromised patients in endemic regions such as Southeast Asia. Improving the accuracy of diagnosing this disease would aid in reducing the mortality of associated infections. In this study, we developed a stable and reproducible murine pulmonary model that mimics human penicilliosis marneffei using a nebulizer to deliver Talaromyces marneffei (SUMS0152) conidia to the lungs of BALB/c nude mice housed in exposure chamber. Using this model, we further revealed that nested PCR was sensitive and specific for detecting Talaromyces marneffei in bronchoalveolar lavage fluid and fresh tissues. This inhalation model may provide a more representative analysis tool for studying the development of penicilliosis marneffei, in addition to revealing that nested PCR has a predictive value in reflecting pulmonary infection.
Collapse
Affiliation(s)
- Yinghui Liu
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaowen Huang
- Department of Dermatology, General Hospital of Guangzhou Military Command of PLA, Guangzhou, China
| | - Xiuwen Yi
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ya He
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Eleftherios Mylonakis
- Division of Infectious Disease, Rhode Island Hospital, Waren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Liyan Xi
- Department of Dermatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- * E-mail:
| |
Collapse
|
47
|
Poczobutt JM, De S, Yadav VK, Nguyen TT, Li H, Sippel TR, Weiser-Evans MCM, Nemenoff RA. Expression Profiling of Macrophages Reveals Multiple Populations with Distinct Biological Roles in an Immunocompetent Orthotopic Model of Lung Cancer. THE JOURNAL OF IMMUNOLOGY 2016; 196:2847-59. [PMID: 26873985 DOI: 10.4049/jimmunol.1502364] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 01/07/2016] [Indexed: 12/13/2022]
Abstract
Macrophages represent an important component of the tumor microenvironment and play a complex role in cancer progression. These cells are characterized by a high degree of plasticity, and they alter their phenotype in response to local environmental cues. Whereas the M1/M2 classification of macrophages has been widely used, the complexity of macrophage phenotypes has not been well studied, particularly in lung cancer. In this study we employed an orthotopic immunocompetent model of lung adenocarcinoma in which murine lung cancer cells are directly implanted into the left lobe of syngeneic mice. Using multimarker flow cytometry, we defined and recovered several distinct populations of monocytes/macrophages from tumors at different stages of progression. We used RNA-seq transcriptional profiling to define distinct features of each population and determine how they change during tumor progression. We defined an alveolar resident macrophage population that does not change in number and expresses multiple genes related to lipid metabolism and lipid signaling. We also defined a population of tumor-associated macrophages that increase dramatically with tumor and selectively expresses a panel of chemokine genes. A third population, which resembles tumor-associated monocytes, expresses a large number of genes involved in matrix remodeling. By correlating transcriptional profiles with clinically prognostic genes, we show that specific monocyte/macrophage populations are enriched in genes that predict outcomes in lung adenocarcinoma, implicating these subpopulations as critical determinants of patient survival. Our data underscore the complexity of monocytes/macrophages in the tumor microenvironment, and they suggest that distinct populations play specific roles in tumor progression.
Collapse
Affiliation(s)
| | - Subhajyoti De
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Vinod K Yadav
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Teresa T Nguyen
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Howard Li
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045; Veterans Affairs Medical Center, Denver, CO 80220; and
| | - Trisha R Sippel
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045
| | - Mary C M Weiser-Evans
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045; Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| | - Raphael A Nemenoff
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045; Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045
| |
Collapse
|
48
|
Duan M, Steinfort DP, Smallwood D, Hew M, Chen W, Ernst M, Irving LB, Anderson GP, Hibbs ML. CD11b immunophenotyping identifies inflammatory profiles in the mouse and human lungs. Mucosal Immunol 2016; 9:550-63. [PMID: 26422753 PMCID: PMC7101582 DOI: 10.1038/mi.2015.84] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 07/12/2015] [Indexed: 02/04/2023]
Abstract
The development of easily accessible tools for human immunophenotyping to classify patients into discrete disease endotypes is advancing personalized therapy. However, no systematic approach has been developed for the study of inflammatory lung diseases with often complex and highly heterogeneous disease etiologies. We have devised an internally standardized flow cytometry approach that can identify parallel inflammatory alveolar macrophage phenotypes in both the mouse and human lungs. In mice, lung innate immune cell alterations during endotoxin challenge, influenza virus infection, and in two genetic models of chronic obstructive lung disease could be segregated based on the presence or absence of CD11b alveolar macrophage upregulation and lung eosinophilia. Additionally, heightened alveolar macrophage CD11b expression was a novel feature of acute lung exacerbations in the SHIP-1(-/-) model of chronic obstructive lung disease, and anti-CD11b antibody administration selectively blocked inflammatory CD11b(pos) but not homeostatic CD11b(neg) alveolar macrophages in vivo. The identification of analogous profiles in respiratory disease patients highlights this approach as a translational avenue for lung disease endotyping and suggests that heterogeneous innate immune cell phenotypes are an underappreciated component of the human lung disease microenvironment.
Collapse
Affiliation(s)
- M Duan
- grid.1002.30000 0004 1936 7857Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Australia ,grid.1008.90000 0001 2179 088XDepartment of Surgery, University of Melbourne, Melbourne, Australia ,grid.482095.2Ludwig Institute for Cancer Research, Melbourne, Australia ,grid.1018.80000 0001 2342 0938Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - D P Steinfort
- grid.416153.40000 0004 0624 1200Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, Australia
| | - D Smallwood
- grid.416153.40000 0004 0624 1200Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, Australia
| | - M Hew
- grid.1623.60000 0004 0432 511XDepartment of Allergy, Immunology and Respiratory Medicine, Alfred Hospital, Parkville, Australia
| | - W Chen
- grid.1018.80000 0001 2342 0938Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, Australia
| | - M Ernst
- grid.1042.7The Walter and Eliza Hall Institute for Medical Research, Parkville, Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, University of Melbourne, Melbourne, Australia ,grid.410678.cPresent Address: 10Present address: Olivia Newton-John Cancer Research Institute, Austin Health, Heidelberg, Victoria 3084, Australia., ,
| | - L B Irving
- grid.416153.40000 0004 0624 1200Department of Respiratory Medicine, Royal Melbourne Hospital, Parkville, Australia
| | - G P Anderson
- grid.1008.90000 0001 2179 088XDepartment of Pharmacology, University of Melbourne, Melbourne, Australia
| | - M L Hibbs
- grid.1002.30000 0004 1936 7857Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Australia
| |
Collapse
|
49
|
Diener KR, Robertson SA, Hayball JD, Lousberg EL. Multi-parameter flow cytometric analysis of uterine immune cell fluctuations over the murine estrous cycle. J Reprod Immunol 2015; 113:61-7. [PMID: 26759962 DOI: 10.1016/j.jri.2015.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/22/2015] [Accepted: 11/30/2015] [Indexed: 01/21/2023]
Abstract
Investigating immune cell populations within various reproductive tissues commonly utilises flow cytometric methods. With advances in fluorophore technology and equipment capabilities, multiple cell types from a single tissue sample can be identified by using different combinations of cell surface markers to distinguish specific cell populations. Here a protocol optimized for mouse uterine tissue was used to show the proportional changes in dendritic cells, monocyte/macrophages, T and B cells, NK and NK T cells, and the granulocytes, neutrophils and eosinophils at each of the four stages of the estrous cycle. Importantly, we demonstrate that use of anti-SiglecF or assessment of FSC/SSC plots could be used to differentiate monocyte/macrophage and eosinophil populations that otherwise cannot be distinguished by use of the common combination of antibodies against F4/80 and CD11b. Our results clearly indicate that within the uterus a dynamic population of immune cells resides, with many cell types reaching peak abundance at estrus and metestrus phases of the cycle, consistent with their importance in the response to paternal antigens and/or pathogens encountered after insemination.
Collapse
Affiliation(s)
- Kerrilyn R Diener
- Experimental Therapeutics Laboratory, Hanson Institute, Royal Adelaide Hospital, and Sansom Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, S.A., 5000, Australia; Robinson Research Institute, Discipline of Obstetrics and Gynaecology, School of Medicine, University of Adelaide, Adelaide, S.A., 5005, Australia.
| | - Sarah A Robertson
- Robinson Research Institute, Discipline of Obstetrics and Gynaecology, School of Medicine, University of Adelaide, Adelaide, S.A., 5005, Australia
| | - John D Hayball
- Experimental Therapeutics Laboratory, Hanson Institute, Royal Adelaide Hospital, and Sansom Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, S.A., 5000, Australia; Robinson Research Institute, Discipline of Obstetrics and Gynaecology, School of Medicine, University of Adelaide, Adelaide, S.A., 5005, Australia
| | - Erin L Lousberg
- Experimental Therapeutics Laboratory, Hanson Institute, Royal Adelaide Hospital, and Sansom Institute, School of Pharmacy and Medical Science, University of South Australia, Adelaide, S.A., 5000, Australia; Robinson Research Institute, Discipline of Obstetrics and Gynaecology, School of Medicine, University of Adelaide, Adelaide, S.A., 5005, Australia
| |
Collapse
|
50
|
Sharma P, Sharma A, Vishwakarma AL, Agnihotri PK, Sharma S, Srivastava M. Host lung immunity is severely compromised during tropical pulmonary eosinophilia: role of lung eosinophils and macrophages. J Leukoc Biol 2015; 99:619-28. [PMID: 26489428 DOI: 10.1189/jlb.4a0715-309rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 10/01/2015] [Indexed: 12/28/2022] Open
Abstract
Eosinophils play a central role in the pathogenesis of tropical pulmonary eosinophilia, a rare, but fatal, manifestation of filariasis. However, no exhaustive study has been done to identify the genes and proteins of eosinophils involved in the pathogenesis of tropical pulmonary eosinophilia. In the present study, we established a mouse model of tropical pulmonary eosinophilia that mimicked filarial manifestations of human tropical pulmonary eosinophilia pathogenesis and used flow cytometry-assisted cell sorting and real-time RT-PCR to study the gene expression profile of flow-sorted, lung eosinophils and lung macrophages during tropical pulmonary eosinophilia pathogenesis. Our results show that tropical pulmonary eosinophilia mice exhibited increased levels of IL-4, IL-5, CCL5, and CCL11 in the bronchoalveolar lavage fluid and lung parenchyma along with elevated titers of IgE and IgG subtypes in the serum. Alveolar macrophages from tropical pulmonary eosinophilia mice displayed decreased phagocytosis, attenuated nitric oxide production, and reduced T-cell proliferation capacity, and FACS-sorted lung eosinophils from tropical pulmonary eosinophilia mice upregulated transcript levels of ficolin A and anti-apoptotic gene Bcl2,but proapoptotic genes Bim and Bax were downregulated. Similarly, flow-sorted lung macrophages upregulated transcript levels of TLR-2, TLR-6, arginase-1, Ym-1, and FIZZ-1 but downregulated nitric oxide synthase-2 levels, signifying their alternative activation. Taken together, we show that the pathogenesis of tropical pulmonary eosinophilia is marked by functional impairment of alveolar macrophages, alternative activation of lung macrophages, and upregulation of anti-apoptotic genes by eosinophils. These events combine together to cause severe lung inflammation and compromised lung immunity. Therapeutic interventions that can boost host immune response in the lungs might thus provide relief to patients with tropical pulmonary eosinophilia.
Collapse
Affiliation(s)
- Pankaj Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Aditi Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Achchhe Lal Vishwakarma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Promod Kumar Agnihotri
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Sharad Sharma
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| | - Mrigank Srivastava
- *Parasitology Division, Sophisticated Analytical Instrument Facility, and Toxicology Division, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India; and Academy of Scientific and Innovative Research, New Delhi, India
| |
Collapse
|