1
|
Waddell LA, Wu Z, Sauter KA, Hope JC, Hume DA. A novel monoclonal antibody against porcine macrophage colony-stimulating factor (CSF1) detects expression on the cell surface of macrophages. Vet Immunol Immunopathol 2023; 266:110681. [PMID: 37992576 DOI: 10.1016/j.vetimm.2023.110681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/24/2023]
Abstract
Macrophage colony-stimulating factor (CSF1) controls the proliferation and differentiation of cells of the mononuclear phagocyte system through binding to the receptor CSF1R. The expression and function of CSF1 has been well-studied in rodents and humans, but knowledge is lacking in other veterinary species. The development of a novel mouse anti-porcine CSF1 monoclonal antibody (mAb) facilitates the characterisation of this growth factor in pigs. Cell surface expression of CSF1 was confirmed on differentiated macrophage populations derived from blood and bone marrow monocytes, and on lung resident macrophages, the first species for this to be confirmed. However, monocytes isolated from blood and bone marrow lacked CSF1 expression. This species-specific mAb delivers the opportunity to further understanding of porcine myeloid cell biology. This is not only vital for the role of pigs as a model for human health, but also as a veterinary species of significant economic and agricultural importance.
Collapse
Affiliation(s)
- Lindsey A Waddell
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Zhiguang Wu
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Kristin A Sauter
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Jayne C Hope
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK.
| | - David A Hume
- Mater Research Institute-University of Queensland, 37 Kent St, Woolloongabba, Qld 4104, Australia
| |
Collapse
|
2
|
Curson JE, Liu L, Luo L, Muusse TW, Lucas RM, Gunther KS, Vajjhala PR, Abrol R, Jones A, Kapetanovic R, Stacey KJ, Stow JL, Sweet MJ. TLR4 phosphorylation at tyrosine 672 activates the ERK/c-FOS signaling module for LPS-induced cytokine responses in macrophages. Eur J Immunol 2023; 53:e2250056. [PMID: 37058370 PMCID: PMC10947571 DOI: 10.1002/eji.202250056] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 03/20/2023] [Accepted: 04/11/2023] [Indexed: 04/15/2023]
Abstract
TLRs engage numerous adaptor proteins and signaling molecules, enabling a complex series of post-translational modifications (PTMs) to mount inflammatory responses. TLRs themselves are post-translationally modified following ligand-induced activation, with this being required to relay the full spectrum of proinflammatory signaling responses. Here, we reveal indispensable roles for TLR4 Y672 and Y749 phosphorylation in mounting optimal LPS-inducible inflammatory responses in primary mouse macrophages. LPS promotes phosphorylation at both tyrosine residues, with Y749 phosphorylation being required for maintenance of total TLR4 protein levels and Y672 phosphorylation exerting its pro-inflammatory effects more selectively by initiating ERK1/2 and c-FOS phosphorylation. Our data also support a role for the TLR4-interacting membrane proteins SCIMP and the SYK kinase axis in mediating TLR4 Y672 phosphorylation to permit downstream inflammatory responses in murine macrophages. The corresponding residue in human TLR4 (Y674) is also required for optimal LPS signaling responses. Our study, thus, reveals how a single PTM on one of the most widely studied innate immune receptors orchestrates downstream inflammatory responses.
Collapse
Affiliation(s)
- James E.B. Curson
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Liping Liu
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Lin Luo
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Timothy W. Muusse
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Richard M. Lucas
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Kimberley S. Gunther
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Parimala R. Vajjhala
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Rishika Abrol
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Alun Jones
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Ronan Kapetanovic
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Katryn J. Stacey
- School of Chemistry and Molecular Biosciences (SCMB) and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Jennifer L. Stow
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| | - Matthew J. Sweet
- Institute for Molecular Bioscience (IMB)IMB Centre for Inflammation and Disease Research and Australian Infectious Diseases Research CentreThe University of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
3
|
Kang S, Berger T. Macrophages in juvenile pig testis: Link with increase in Sertoli cells induced by oestradiol suppression. Reprod Domest Anim 2023; 58:564-568. [PMID: 36645769 DOI: 10.1111/rda.14322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/25/2022] [Accepted: 12/14/2022] [Indexed: 01/17/2023]
Abstract
Macrophage presence and location were evaluated in juvenile boar testes at the end of the first wave of Sertoli cell proliferation. Macrophage presence was compared in littermate boars treated with letrozole, a treatment which extended this first wave of proliferation beyond the sampling timepoint. Macrophages were identified as the CD68 positive cells following immunohistochemical labelling of paraffin sections and parenchymal macrophages enumerated. Macrophages present in a layer beneath the tunica albuginea received a score based on density and thickness of this layer. Density within the testicular parenchyma was highly variable in vehicle-treated boars (>100-fold) and did not differ from that observed in the letrozole-treated littermates. However, the macrophage layer beneath the tunica albuginea was denser and thicker in the letrozole-treated animals than in their vehicle-treated littermates. This suggests that macrophages might be involved in the letrozole-induced prolongation of Sertoli cell proliferation.
Collapse
Affiliation(s)
- Soyeon Kang
- Department of Animal Science, University of California, Davis, Davis, California, USA
| | - Trish Berger
- Department of Animal Science, University of California, Davis, Davis, California, USA
| |
Collapse
|
4
|
Chandra J, Teoh SM, Kuo P, Tolley L, Bashaw AA, Tuong ZK, Liu Y, Chen Z, Wells JW, Yu C, Frazer IH, Yu M. Manganese-Doped Silica-Based Nanoparticles Promote the Efficacy of Antigen-Specific Immunotherapy. THE JOURNAL OF IMMUNOLOGY 2021; 206:987-998. [PMID: 33504616 DOI: 10.4049/jimmunol.2000355] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022]
Abstract
Prophylactic human papillomavirus (HPV) vaccines are commercially available for prevention of infection with cancerogenic HPV genotypes but are not able to combat pre-existing HPV-associated disease. In this study, we designed a nanomaterial-based therapeutic HPV vaccine, comprising manganese (Mn4+)-doped silica nanoparticles (Mn4+-SNPs) and the viral neoantigen peptide GF001 derived from the HPV16 E7 oncoprotein. We show in mice that Mn4+-SNPs act as self-adjuvants by activating the inflammatory signaling pathway via generation of reactive oxygen species, resulting in immune cell recruitment to the immunization site and dendritic cell maturation. Mn4+-SNPs further serve as Ag carriers by facilitating endo/lysosomal escape via depletion of protons in acidic endocytic compartments and subsequent Ag delivery to the cytosol for cross-presentation. The Mn4+-SNPs+GF001 nanovaccine induced strong E7-specific CD8+ T cell responses, leading to remission of established murine HPV16 E7-expressing solid TC-1 tumors and E7-expressing transgenic skin grafts. This vaccine construct offers a simple and general strategy for therapeutic HPV and potentially other cancer vaccines.
Collapse
Affiliation(s)
- Janin Chandra
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Siok Min Teoh
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Paula Kuo
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Lynn Tolley
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Abate Assefa Bashaw
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Zewen Kelvin Tuong
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Yang Liu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; and
| | - Zibin Chen
- Australian Centre for Microscopy and Microanalysis, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - James W Wells
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; and
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia;
| | - Meihua Yu
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland 4102, Australia; .,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia; and
| |
Collapse
|
5
|
Karagianni AE, Lisowski ZM, Hume DA, Scott Pirie R. The equine mononuclear phagocyte system: The relevance of the horse as a model for understanding human innate immunity. Equine Vet J 2020; 53:231-249. [PMID: 32881079 DOI: 10.1111/evj.13341] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/07/2020] [Accepted: 08/13/2020] [Indexed: 12/11/2022]
Abstract
The mononuclear phagocyte system (MPS) is a family of cells of related function that includes bone marrow progenitors, blood monocytes and resident tissue macrophages. Macrophages are effector cells in both innate and acquired immunity. They are a major resident cell population in every organ and their numbers increase in response to proinflammatory stimuli. Their function is highly regulated by a wide range of agonists, including lymphokines, cytokines and products of microorganisms. Macrophage biology has been studied most extensively in mice, yet direct comparisons of rodent and human macrophages have revealed many functional differences. In this review, we provide an overview of the equine MPS, describing the variation in the function and phenotype of macrophages depending on their location and the similarities and differences between the rodent, human and equine immune response. We discuss the use of the horse as a large animal model in which to study macrophage biology and pathological processes shared with humans. Finally, following the recent update to the horse genome, facilitating further comparative analysis of regulated gene expression between the species, we highlight the importance of future transcriptomic macrophage studies in the horse, the findings of which may also be applicable to human as well as veterinary research.
Collapse
Affiliation(s)
- Anna E Karagianni
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Zofia M Lisowski
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - David A Hume
- Mater Research Institute-UQ, Translational Research Institute, Woolloongabba, QLD, Australia
| | - R Scott Pirie
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
6
|
Bush SJ, McCulloch MEB, Lisowski ZM, Muriuki C, Clark EL, Young R, Pridans C, Prendergast JGD, Summers KM, Hume DA. Species-Specificity of Transcriptional Regulation and the Response to Lipopolysaccharide in Mammalian Macrophages. Front Cell Dev Biol 2020; 8:661. [PMID: 32793601 PMCID: PMC7386301 DOI: 10.3389/fcell.2020.00661] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/01/2020] [Indexed: 02/02/2023] Open
Abstract
Mammalian macrophages differ in their basal gene expression profiles and response to the toll-like receptor 4 (TLR4) agonist, lipopolysaccharide (LPS). In human macrophages, LPS elicits a temporal cascade of transient gene expression including feed forward activators and feedback regulators that limit the response. Here we present a transcriptional network analysis of the response of sheep bone marrow-derived macrophages (BMDM) to LPS based upon RNA-seq at 0, 2, 4, 7, and 24 h post-stimulation. The analysis reveals a conserved transcription factor network with humans, and rapid induction of feedback regulators that constrain the response at every level. The gene expression profiles of sheep BMDM at 0 and 7 h post LPS addition were compared to similar data obtained from goat, cow, water buffalo, horse, pig, mouse and rat BMDM. This comparison was based upon identification of 8,200 genes annotated in all species and detected at >10TPM in at least one sample. Analysis of expression of transcription factors revealed a conserved transcriptional millieu associated with macrophage differentiation and LPS response. The largest co-expression clusters, including genes encoding cell surface receptors, endosome–lysosome components and secretory activity, were also expressed in all species and the combined dataset defines a macrophage functional transcriptome. All of the large animals differed from rodents in lacking inducible expression of genes involved in arginine metabolism and nitric oxide production. Instead, they expressed inducible transporters and enzymes of tryptophan and kynurenine metabolism. BMDM from all species expressed high levels of transcripts encoding transporters and enzymes involved in glutamine metabolism suggesting that glutamine is a major metabolic fuel. We identify and discuss transcripts that were uniquely expressed or regulated in rodents compared to large animals including ACOD1, CXC and CC chemokines, CD163, CLEC4E, CPM, CSF1, CSF2, CTSK, MARCO, MMP9, SLC2A3, SLC7A7, and SUCNR1. Conversely, the data confirm the conserved regulation of multiple transcripts for which there is limited functional data from mouse models and knockouts. The data provide a resource for functional annotation and interpretation of loci involved in susceptibility to infectious and inflammatory disease in humans and large animal species.
Collapse
Affiliation(s)
- Stephen J Bush
- Nuffield Department of Clinical Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Mary E B McCulloch
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Zofia M Lisowski
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Charity Muriuki
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Emily L Clark
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Rachel Young
- The Roslin Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Clare Pridans
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom.,Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | | | - Kim M Summers
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| | - David A Hume
- Mater Research Institute-University of Queensland, Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
7
|
Hume DA, Caruso M, Ferrari-Cestari M, Summers KM, Pridans C, Irvine KM. Phenotypic impacts of CSF1R deficiencies in humans and model organisms. J Leukoc Biol 2019; 107:205-219. [PMID: 31330095 DOI: 10.1002/jlb.mr0519-143r] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/20/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022] Open
Abstract
Mϕ proliferation, differentiation, and survival are controlled by signals from the Mϕ CSF receptor (CSF1R). Mono-allelic gain-of-function mutations in CSF1R in humans are associated with an autosomal-dominant leukodystrophy and bi-allelic loss-of-function mutations with recessive skeletal dysplasia, brain disorders, and developmental anomalies. Most of the phenotypes observed in these human disease states are also observed in mice and rats with loss-of-function mutations in Csf1r or in Csf1 encoding one of its two ligands. Studies in rodent models also highlight the importance of genetic background and likely epistatic interactions between Csf1r and other loci. The impacts of Csf1r mutations on the brain are usually attributed solely to direct impacts on microglial number and function. However, analysis of hypomorphic Csf1r mutants in mice and several other lines of evidence suggest that primary hydrocephalus and loss of the physiological functions of Mϕs in the periphery contribute to the development of brain pathology. In this review, we outline the evidence that CSF1R is expressed exclusively in mononuclear phagocytes and explore the mechanisms linking CSF1R mutations to pleiotropic impacts on postnatal growth and development.
Collapse
Affiliation(s)
- David A Hume
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Melanie Caruso
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | | | - Kim M Summers
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Clare Pridans
- Centre for Inflammation Research, The University of Edinburgh, Edinburgh, United Kingdom
| | - Katharine M Irvine
- Mater Research Institute, University of Queensland, Woolloongabba, Queensland, Australia
| |
Collapse
|
8
|
Novel function of PiT1/SLC20A1 in LPS-related inflammation and wound healing. Sci Rep 2019; 9:1808. [PMID: 30755642 PMCID: PMC6372663 DOI: 10.1038/s41598-018-37551-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/27/2018] [Indexed: 12/17/2022] Open
Abstract
PiT1/SLC20A1 is an inorganic phosphate transporter with additional functions including the regulation of TNFα-induced apoptosis, erythropoiesis, cell proliferation and insulin signaling. Recent data suggest a relationship between PiT1 and NF-κB-dependent inflammation: (i) Pit1 mRNA is up-regulated in the context of NF-κB pathway activation; (ii) NF-κB target gene transcription is decreased in PiT1-deficient conditions. This led us to investigate the role of PiT1 in lipopolysaccharide (LPS)-induced inflammation. MCP-1 and IL-6 concentrations were impaired in PiT1-deficient bone marrow derived macrophages (BMDMs) upon LPS stimulation. Lower MCP-1 and IL-6 serum levels were observed in Mx1-Cre; Pit1lox/lox mice dosed intraperitoneally with LPS. Lower PiT1 expression correlated with decreased in vitro wound healing and lower reactive oxygen species levels. Reduced IκB degradation and lower p65 nuclear translocation were observed in PiT1-deficient cells stimulated with LPS. Conversely, PiT1 expression was induced in vitro upon LPS stimulation. Addition of an NF-κB inhibitor abolished LPS-induced PiT1 expression. Furthermore, we showed that p65 expression activated Pit1 promoter activity. Finally, ChIP assays demonstrated that p65 directly binds to the mPit1 promoter in response to LPS. These data demonstrate a completely novel function of PiT1 in the response to LPS and provide mechanistic insights into the regulation of PiT1 expression by NF-κB.
Collapse
|
9
|
Kurena B, Müller E, Christopoulos PF, Johnsen IB, Stankovic B, Øynebråten I, Corthay A, Zajakina A. Generation and Functional In Vitro Analysis of Semliki Forest Virus Vectors Encoding TNF-α and IFN-γ. Front Immunol 2017; 8:1667. [PMID: 29276511 PMCID: PMC5727424 DOI: 10.3389/fimmu.2017.01667] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/14/2017] [Indexed: 12/25/2022] Open
Abstract
Cytokine gene delivery by viral vectors is a promising novel strategy for cancer immunotherapy. Semliki Forest virus (SFV) has many advantages as a delivery vector, including the ability to (i) induce p53-independent killing of tumor cells via apoptosis, (ii) elicit a type-I interferon (IFN) response, and (iii) express high levels of the transgene. SFV vectors encoding cytokines such as interleukin (IL)-12 have shown promising therapeutic responses in experimental tumor models. Here, we developed two new recombinant SFV vectors encoding either murine tumor necrosis factor-α (TNF-α) or murine interferon-γ (IFN-γ), two cytokines with documented immunostimulatory and antitumor activity. The SFV vector showed high infection rate and cytotoxicity in mouse and human lung carcinoma cells in vitro. By contrast, mouse and human macrophages were resistant to infection with SFV. The recombinant SFV vectors directly inhibited mouse lung carcinoma cell growth in vitro, while exploiting the cancer cells for production of SFV vector-encoded cytokines. The functionality of SFV vector-derived TNF-α was confirmed through successful induction of cell death in TNF-α-sensitive fibroblasts in a concentration-dependent manner. SFV vector-derived IFN-γ activated macrophages toward a tumoricidal phenotype leading to suppressed Lewis lung carcinoma cell growth in vitro in a concentration-dependent manner. The ability of SFV to provide functional cytokines and infect tumor cells but not macrophages suggests that SFV may be very useful for cancer immunotherapy employing tumor-infiltrating macrophages.
Collapse
Affiliation(s)
- Baiba Kurena
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway.,Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Elisabeth Müller
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Panagiotis F Christopoulos
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Ingvild Bjellmo Johnsen
- Department of Laboratory Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Branislava Stankovic
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Inger Øynebråten
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Alexandre Corthay
- Tumor Immunology Lab, Department of Pathology, Rikshospitalet, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Anna Zajakina
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Riga, Latvia
| |
Collapse
|
10
|
Gil-Pulido J, Cochain C, Lippert MA, Schneider N, Butt E, Amézaga N, Zernecke A. Deletion of Batf3-dependent antigen-presenting cells does not affect atherosclerotic lesion formation in mice. PLoS One 2017; 12:e0181947. [PMID: 28771609 PMCID: PMC5542449 DOI: 10.1371/journal.pone.0181947] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/10/2017] [Indexed: 02/02/2023] Open
Abstract
Atherosclerosis is the main underlying cause for cardiovascular events such as myocardial infarction and stroke and its development might be influenced by immune cells. Dendritic cells (DCs) bridge innate and adaptive immune responses by presenting antigens to T cells and releasing a variety of cytokines. Several subsets of DCs can be discriminated that engage specific transcriptional pathways for their development. Basic leucine zipper transcription factor ATF-like 3 (Batf3) is required for the development of classical CD8α+ and CD103+ DCs. By crossing mice deficient in Batf3 with atherosclerosis-prone low density lipoprotein receptor (Ldlr-/-)-deficient mice we here aimed to further address the contribution of Batf3-dependent CD8α+ and CD103+ antigen-presenting cells to atherosclerosis. We demonstrate that deficiency in Batf3 entailed mild effects on the immune response in the spleen but did not alter atherosclerotic lesion formation in the aorta or aortic root, nor affected plaque phenotype in low density lipoprotein receptor-deficient mice fed a high fat diet. We thus provide evidence that Batf3-dependent antigen-presenting cells do not have a prominent role in atherosclerosis.
Collapse
Affiliation(s)
- Jesus Gil-Pulido
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Clement Cochain
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Malte A. Lippert
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Nicole Schneider
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Elke Butt
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Núria Amézaga
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Alma Zernecke
- Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
- * E-mail:
| |
Collapse
|
11
|
SCIMP is a transmembrane non-TIR TLR adaptor that promotes proinflammatory cytokine production from macrophages. Nat Commun 2017; 8:14133. [PMID: 28098138 PMCID: PMC5253658 DOI: 10.1038/ncomms14133] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/02/2016] [Indexed: 01/04/2023] Open
Abstract
Danger signals activate Toll-like receptors (TLRs), thereby initiating inflammatory responses. Canonical TLR signalling, via Toll/Interleukin-1 receptor domain (TIR)-containing adaptors and proinflammatory transcription factors such as NF-κB, occurs in many cell types; however, additional mechanisms are required for specificity of inflammatory responses in innate immune cells. Here we show that SCIMP, an immune-restricted, transmembrane adaptor protein (TRAP), promotes selective proinflammatory cytokine responses by direct modulation of TLR4. SCIMP is a non-TIR-containing adaptor, binding directly to the TLR4-TIR domain in response to lipopolysaccharide. In macrophages, SCIMP is constitutively associated with the Lyn tyrosine kinase, is required for tyrosine phosphorylation of TLR4, and facilitates TLR-inducible production of the proinflammatory cytokines IL-6 and IL-12p40. Point mutations in SCIMP abrogating TLR4 binding also prevent SCIMP-mediated cytokine production. SCIMP is, therefore, an immune-specific TLR adaptor that shapes host defence and inflammation. Toll-like receptors engage TIR domain-containing adaptors to control proinflammatory gene expression in response to pathogens and tissue damage. Here the authors show that the non-TIR domain-containing transmembrane protein SCIMP is a previously unrecognized TLR adaptor expressed by macrophages.
Collapse
|
12
|
Xu X, Yuan X, Li N, Dewey WL, Li PL, Zhang F. Lysosomal cholesterol accumulation in macrophages leading to coronary atherosclerosis in CD38(-/-) mice. J Cell Mol Med 2016; 20:1001-13. [PMID: 26818887 PMCID: PMC4882979 DOI: 10.1111/jcmm.12788] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 12/13/2015] [Indexed: 12/16/2022] Open
Abstract
The disruption in transportation of oxLDL‐derived cholesterol and the subsequent lipid accumulation in macrophages are the hallmark events in atherogenesis. Our recent studies demonstrated that lysosomal Ca2+ messenger of nicotinic acid adenine dinucleotide phosphate (NAADP), an enzymatic product of CD38 ADP‐ribosylcyclase (CD38), promoted lipid endocytic trafficking in human fibroblast cells. The current studies are designed to examine the functional role of CD38/NAADP pathway in the regulation of lysosomal cholesterol efflux in atherosclerosis. Oil red O staining showed that oxLDL concentration‐dependently increased lipid buildup in bone marrow‐derived macrophages from both wild type and CD38−/−, but to a significant higher extent with CD38 gene deletion. Bodipy 493/503 fluorescence staining found that the deposited lipid in macrophages was mainly enclosed in lysosomal organelles and largely enhanced with the blockade of CD38/NAADP pathway. Filipin staining and direct measurement of lysosome fraction further revealed that the free cholesterol constituted a major portion of the total cholesterol segregated in lysosomes. Moreover, in situ assay disclosed that both lysosomal lumen acidity and the acid lipase activity were reduced upon cholesterol buildup in lysosomes. In CD38−/− mice, treatment with Western diet (12 weeks) produced atherosclerotic damage in coronary artery with striking lysosomal cholesterol sequestration in macrophages. These data provide the first experimental evidence that the proper function of CD38/NAADP pathway plays an essential role in promoting free cholesterol efflux from lysosomes and that a defection of this signalling leads to lysosomal cholesterol accumulation in macrophages and results in coronary atherosclerosis in CD38−/− mice.
Collapse
Affiliation(s)
- Xiaoyang Xu
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Xinxu Yuan
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - William L Dewey
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| | - Fan Zhang
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
13
|
Tuong ZK, Lau P, Du X, Condon ND, Goode JM, Oh TG, Yeo JC, Muscat GEO, Stow JL. RORα and 25-Hydroxycholesterol Crosstalk Regulates Lipid Droplet Homeostasis in Macrophages. PLoS One 2016; 11:e0147179. [PMID: 26812621 PMCID: PMC4727927 DOI: 10.1371/journal.pone.0147179] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 12/30/2015] [Indexed: 11/19/2022] Open
Abstract
Nuclear hormone receptors have important roles in the regulation of metabolic and inflammatory pathways. The retinoid-related orphan receptor alpha (Rorα)-deficient staggerer (sg/sg) mice display several phenotypes indicative of aberrant lipid metabolism, including dyslipidemia, and increased susceptibility to atherosclerosis. In this study we demonstrate that macrophages from sg/sg mice have increased ability to accumulate lipids and accordingly exhibit larger lipid droplets (LD). We have previously shown that BMMs from sg/sg mice have significantly decreased expression of cholesterol 25-hydroxylase (Ch25h) mRNA, the enzyme that produces the oxysterol, 25-hydroxycholesterol (25HC), and now confirm this at the protein level. 25HC functions as an inverse agonist for RORα. siRNA knockdown of Ch25h in macrophages up-regulates Vldlr mRNA expression and causes increased accumulation of LDs. Treatment with physiological concentrations of 25HC in sg/sg macrophages restored lipid accumulation back to normal levels. Thus, 25HC and RORα signify a new pathway involved in the regulation of lipid homeostasis in macrophages, potentially via increased uptake of lipid which is suggested by mRNA expression changes in Vldlr and other related genes.
Collapse
MESH Headings
- Animals
- Bone Marrow Cells/cytology
- Cells, Cultured
- Chromatography, Thin Layer
- Drug Inverse Agonism
- Hydroxycholesterols/metabolism
- Lipid Droplets/metabolism
- Lipid Metabolism
- Lipids/analysis
- Macrophages/cytology
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C3H
- Mice, Knockout
- Nuclear Receptor Subfamily 1, Group F, Member 1/chemistry
- Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism
- RNA Interference
- RNA, Messenger/metabolism
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Steroid Hydroxylases/antagonists & inhibitors
- Steroid Hydroxylases/genetics
- Steroid Hydroxylases/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Zewen Kelvin Tuong
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Patrick Lau
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Ximing Du
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney NSW 2052, Australia
| | - Nicholas D. Condon
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Joel M. Goode
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Tae Gyu Oh
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - Jeremy C. Yeo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
| | - George E. O. Muscat
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
- * E-mail: (GEOM); (JLS)
| | - Jennifer L. Stow
- Institute for Molecular Bioscience, The University of Queensland, Brisbane QLD 4072, Australia
- * E-mail: (GEOM); (JLS)
| |
Collapse
|
14
|
Ravasi T, Mavromatis CH, Bokil NJ, Schembri MA, Sweet MJ. Co-transcriptomic Analysis by RNA Sequencing to Simultaneously Measure Regulated Gene Expression in Host and Bacterial Pathogen. Methods Mol Biol 2016; 1390:145-158. [PMID: 26803628 DOI: 10.1007/978-1-4939-3335-8_10] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Intramacrophage pathogens subvert antimicrobial defence pathways using various mechanisms, including the targeting of host TLR-mediated transcriptional responses. Conversely, TLR-inducible host defence mechanisms subject intramacrophage pathogens to stress, thus altering pathogen gene expression programs. Important biological insights can thus be gained through the analysis of gene expression changes in both the host and the pathogen during an infection. Traditionally, research methods have involved the use of qPCR, microarrays and/or RNA sequencing to identify transcriptional changes in either the host or the pathogen. Here we describe the application of RNA sequencing using samples obtained from in vitro infection assays to simultaneously quantify both host and bacterial pathogen gene expression changes, as well as general approaches that can be undertaken to interpret the RNA sequencing data that is generated. These methods can be used to provide insights into host TLR-regulated transcriptional responses to microbial challenge, as well as pathogen subversion mechanisms against such responses.
Collapse
Affiliation(s)
- Timothy Ravasi
- Integrative Systems Biology Laboratory, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia.
- Division of Computer, Electricaland Mathematical Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia.
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Charalampos Harris Mavromatis
- Integrative Systems Biology Laboratory, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
- Division of Computer, Electricaland Mathematical Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
- Division of Medical Genetics, Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Nilesh J Bokil
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
| | - Mark A Schembri
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
- Australian Infectious Diseases Research Centre, The University of Queensland, St Lucia, Brisbane, QLD, 4072, Australia.
| |
Collapse
|
15
|
Maroni D, Rana S, Mukhopadhyay C, Natarajan A, Naramura M. A quinoxaline urea analog uncouples inflammatory and pro-survival functions of IKKβ. Immunol Lett 2015; 168:319-24. [PMID: 26518140 DOI: 10.1016/j.imlet.2015.10.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/15/2015] [Accepted: 10/20/2015] [Indexed: 11/15/2022]
Abstract
Activation of the NF-κB pathway is causally linked to initiation and progression of diverse cancers. Therefore, IKKβ, the key regulatory kinase of the canonical NF-κB pathway, should be a logical target for cancer treatment. However, existing IKKβ inhibitors are known to induce paradoxical immune activation, which limits their clinical usefulness. Recently, we identified a quinoxaline urea analog 13-197 as a novel IKKβ inhibitor that delays tumor growth without significant adverse effects in xenograft tumor models. In the present study, we found that 13-197 had little effect on LPS-induced NF-κB target gene induction by primary mouse macrophages while maintaining considerable anti-proliferative activities. These characteristics may explain absence of inflammatory side effects in animals treated with 13-197. Our data also demonstrate that the inflammation and proliferation-related functions of IKKβ can be uncoupled, and highlight the utility of 13-197 to dissect these downstream pathways.
Collapse
Affiliation(s)
- Dulce Maroni
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Sandeep Rana
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Chandrani Mukhopadhyay
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mayumi Naramura
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
16
|
Halvorsen B, Espeland MZ, Andersen GØ, Yndestad A, Sagen EL, Rashidi A, Knudsen EC, Skjelland M, Skagen KR, Krohg-Sørensen K, Holm S, Ritschel V, Holven KB, Biessen EAL, Aukrust P, Dahl TB. Increased expression of NAMPT in PBMC from patients with acute coronary syndrome and in inflammatory M1 macrophages. Atherosclerosis 2015; 243:204-10. [PMID: 26402139 DOI: 10.1016/j.atherosclerosis.2015.09.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 09/02/2015] [Accepted: 09/02/2015] [Indexed: 10/23/2022]
Abstract
AIM The aim of the present study were to elucidate the role of NAMPT in atherosclerosis, by examine NAMPT expression in peripheral blood mononuclear cells (PBMC) in patients with coronary artery disease (CAD) and healthy controls and by examining the regulation and effect of NAMPT on macrophage polarization, hypothesizing that it could influence the polarization to inflammatory and resolving macrophages. METHOD AND RESULTS We analyzed RNA levels of NAMPT in PBMC from CAD and healthy controls and found NAMPT to be increased in PBMC from patients with acute coronary syndrome (n = 39) compared to healthy controls (n = 20) and patients with stable CAD (n = 22). Within the PBMC NAMPT was correlated to several inflammatory cytokines and the antioxidant enzyme superoxide dismutase 2. In vitro cell experiments revealed that NAMPT is increased both intracellular and extracellular in inflammatory M1 macrophages compared to in anti-inflammatory M2 macrophages. In addition, inhibiting NAMPT enzymatic activity inhibited M1 polarization in macrophages. CONCLUSION Based on our in vivo and in vitro findings we suggest that NAMPT could contribute to systemic and plaque inflammation in atherosclerotic disorders at least partly through effect on macrophages.
Collapse
Affiliation(s)
- Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway
| | - Martine Z Espeland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Geir Øystein Andersen
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Center for Heart Failure, University of Oslo, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway; Center for Heart Failure, University of Oslo, Oslo, Norway
| | - Ellen Lund Sagen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Azita Rashidi
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Eva C Knudsen
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Mona Skjelland
- Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Karolina R Skagen
- Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Kirsten Krohg-Sørensen
- Department of Thoracic and Cardiovascular Surgery Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Vibeke Ritschel
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Center for Clinical Heart Research, Oslo University Hospital Ullevål, Oslo, Norway
| | - Kirsten B Holven
- Department of Nutrition, Institute for Basic Medical Sciences, University of Oslo, Norway
| | - Erik A L Biessen
- Experimental Vascular Pathology Group, Department of Pathology, CARIM, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway; Section of Clinical Immunology and Infectious Disease, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway.
| |
Collapse
|
17
|
Xu X, Zhang A, Li N, Li PL, Zhang F. Concentration-Dependent Diversifcation Effects of Free Cholesterol Loading on Macrophage Viability and Polarization. Cell Physiol Biochem 2015; 37:419-431. [PMID: 26314949 DOI: 10.1159/000430365] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2015] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND/AIMS The accumulation of free cholesterol in atherosclerotic lesions has been well documented in both animals and humans. In studying the relevance of free cholesterol buildup in atherosclerosis, contradictory results have been generated, indicating that free cholesterol produces both pro- and anti-atherosclerosis effects in macrophages. This inconsistency might stem from the examination of only select concentrations of free cholesterol. In the present study, we sought to investigate the implication of excess free cholesterol loading in the pathophysiology of atherosclerosis across a broad concentration range from (in µg/ml) 0 to 60. METHODS Macrophage viability was determined by measuring formazan formation and flow cytometry viable cell counting. The polarization of M1 and M2 macrophages was differentiated by FACS (Fluorescence-Activated Cell Sorting) assay. The secretion of IL-1β in macrophage culture medium was measured by ELISA kit. Macrophage apoptosis was detected by flow cytometry using a TUNEL kit. RESULTS Macrophage viability was increased at the treatment of lower concentrations of free cholesterol from (in µg/ml) 0 to 20, but gradually decreased at higher concentrations from 20 to 60. Lower free cholesterol loading induced anti-inflammatory M2 macrophage polarization. The activation of the PPARx03B3; (Peroxisome Proliferator-Activated Receptor gamma) nuclear factor underscored the stimulation of this M2 phenotype. Nevertheless, higher levels of free cholesterol resulted in pro-inflammatory M1 activation. Moreover, with the application of higher free cholesterol concentrations, macrophage apoptosis and secretion of the inflammatory cytokine IL-1β increased significantly. CONCLUSION These results for the first time demonstrate that free cholesterol could render concentration-dependent diversification effects on macrophage viability, polarization, apoptosis and inflammatory cytokine secretions, thereby reconciling the pros and cons of free cholesterol buildup in macrophages to the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Xiaoyang Xu
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, VA 23298
| | - Aolin Zhang
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, VA 23298
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, VA 23298
| | - Pin-Lan Li
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, VA 23298
| | - Fan Zhang
- Department of Pharmacology & Toxicology, Medical College of Virginia, Virginia Commonwealth University, VA 23298
| |
Collapse
|
18
|
Echogenic perfluorohexane-loaded macrophages adhere in vivo to activated vascular endothelium in mice, an explorative study. Cardiovasc Ultrasound 2015; 13:1. [PMID: 25567641 PMCID: PMC4293794 DOI: 10.1186/1476-7120-13-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 12/23/2014] [Indexed: 01/11/2023] Open
Abstract
Background Macrophages may concentrate ultrasound contrast agents and exhibit selective adhesion to activated endothelium. The present study investigates in mice the potential of perfluorohexane (PFH) loaded macrophages to act as ultrasound contrast agent with high reflectivity and specifically targeted at (atherosclerotic) vascular lesions. Methods Lung passage was evaluated with a mouse echo scanner after injection, at a slow pace or as a bolus, of varying doses of PFH-loaded and unloaded bone marrow macrophages (BMM) into the jugular vein. The interaction of PFH-loaded and unloaded BMM with TNF-α stimulated carotid artery endothelium after tail vein injection was assessed by means of intravital microscopy. Results High doses of jugular vein injected PFH-loaded BMM were visible with ultrasound in the pulmonary artery and detectable in the carotid artery. At intravital microscopy, tail vein injected BMM exhibited rolling and adhesion behavior at the TNF-α stimulated carotid endothelium, similar to that of native blood leukocytes. Rolling behavior was not different between PFH-loaded and unloaded BMM (p = 0.38). Conclusion In vivo, perfluorohexane loaded macrophages pass the pulmonary circulation and appear on the arterial side. Moreover, they roll and adhere selectively to activated endothelium under physiological flow conditions. These findings indicate that perfluorohexane loaded BMM could be used to study processes in vivo where endothelial activation plays a role, such as atherosclerosis.
Collapse
|
19
|
Yeo JC, Wall AA, Stow JL, Hamilton NA. High-throughput quantification of early stages of phagocytosis. Biotechniques 2013; 55:115-24. [DOI: 10.2144/000114075] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Accepted: 07/29/2013] [Indexed: 11/23/2022] Open
Abstract
Phagocytosis—the engulfment of cells and foreign bodies—is an important cellular process in innate immunity, development, and disease. Quantification of various stages of phagocytosis, especially in a rapid screening fashion, is an invaluable tool for elucidating protein function during this process. However, current methods for assessing phagocytosis are largely limited to flow cytometry and manual image-based assays, providing limited information. Here, we present an image-based, semi-automated phagocytosis assay to rapidly quantitate three distinct stages during the early engulfment of opsonized beads. Captured images are analyzed using the image-processing software ImageJ and quantified using a macro. Modifications to this method allowed quantification of phagocytosis only in fluorescently labeled transfected cells. Additionally, the time course of bead internalization could be measured using this approach. The assay could discriminate perturbations to stages of phagocytosis induced by known pharmacological inhibitors of filamentous actin and phosphoinositol-3-kinase. Our methodology offers the ability to automatically categorize large amounts of image data into the three early stages of phagocytosis within minutes, clearly demonstrating its potential value in investigating aberrant phagocytosis when manipulating proteins of interest in drug screens and disease.
Collapse
Affiliation(s)
- Jeremy Changyu Yeo
- Institute for Molecular Bioscience and NHMRC Program in Cellular Microbiology, The University of Queensland, Brisbane, Australia
| | - Adam Alexander Wall
- Institute for Molecular Bioscience and NHMRC Program in Cellular Microbiology, The University of Queensland, Brisbane, Australia
| | - Jennifer Lea Stow
- Institute for Molecular Bioscience and NHMRC Program in Cellular Microbiology, The University of Queensland, Brisbane, Australia
| | - Nicholas Ahti Hamilton
- Institute for Molecular Bioscience and NHMRC Program in Cellular Microbiology, The University of Queensland, Brisbane, Australia
| |
Collapse
|
20
|
Macrophage fusion is controlled by the cytoplasmic protein tyrosine phosphatase PTP-PEST/PTPN12. Mol Cell Biol 2013; 33:2458-69. [PMID: 23589331 DOI: 10.1128/mcb.00197-13] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Macrophages can undergo cell-cell fusion, leading to the formation of multinucleated giant cells and osteoclasts. This process is believed to promote the proteolytic activity of macrophages toward pathogens, foreign bodies, and extracellular matrices. Here, we examined the role of PTP-PEST (PTPN12), a cytoplasmic protein tyrosine phosphatase, in macrophage fusion. Using a macrophage-targeted PTP-PEST-deficient mouse, we determined that PTP-PEST was not needed for macrophage differentiation or cytokine production. However, it was necessary for interleukin-4-induced macrophage fusion into multinucleated giant cells in vitro. It was also needed for macrophage fusion following implantation of a foreign body in vivo. Moreover, in the RAW264.7 macrophage cell line, PTP-PEST was required for receptor activator of nuclear factor kappa-B ligand (RANKL)-triggered macrophage fusion into osteoclasts. PTP-PEST had no impact on expression of fusion mediators such as β-integrins, E-cadherin, and CD47, which enable macrophages to become fusion competent. However, it was needed for polarization of macrophages, migration induced by the chemokine CC chemokine ligand 2 (CCL2), and integrin-induced spreading, three key events in the fusion process. PTP-PEST deficiency resulted in specific hyperphosphorylation of the protein tyrosine kinase Pyk2 and the adaptor paxillin. Moreover, a fusion defect was induced upon treatment of normal macrophages with a Pyk2 inhibitor. Together, these data argue that macrophage fusion is critically dependent on PTP-PEST. This function is seemingly due to the ability of PTP-PEST to control phosphorylation of Pyk2 and paxillin, thereby regulating cell polarization, migration, and spreading.
Collapse
|
21
|
Tuong ZK, Lau P, Yeo JC, Pearen MA, Wall AA, Stanley AC, Stow JL, Muscat GEO. Disruption of Rorα1 and cholesterol 25-hydroxylase expression attenuates phagocytosis in male Rorαsg/sg mice. Endocrinology 2013; 154:140-9. [PMID: 23239817 DOI: 10.1210/en.2012-1889] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We and others have previously demonstrated that congenital deficiency of the nuclear hormone receptor, Rorα1, in staggerer (sg/sg) mice results in resistance to diet-induced obesity and increased insulin sensitivity. Paradoxically, the sg/sg mice are susceptible to atherosclerosis and display impaired innate immunity, underscoring the regulatory links between metabolic disease, inflammation, and susceptibility to infection. Here, we present novel evidence that Rorα1 regulates innate immune function by demonstrating impaired phagocytosis in sg/sg mice. The early stages of Fc-γ receptor-mediated phagocytosis in lipopolysaccharide-activated sg/sg bone marrow-derived macrophages (BMMs) were significantly impaired compared with wild-type cells. Moreover, in sg/sg BMMs, the phagocytic cup membranes had reduced levels of cholesterol. Expression profiling revealed dysregulated expression of genes involved in inflammation and lipid metabolism in sg/sg BMMs. Notably, we identified decreased expression of the mRNA encoding cholesterol 25-hydroxylase (Ch25h), an enzyme that converts cholesterol to 25-hydroxycholesterol (25HC), an oxysterol with emerging roles in immunity. Treatment of sg/sg BMMs with 25HC rescued phagocytosis in a dose-dependent manner, whereas small interfering RNA knockdown of Ch25h mRNA expression in wild-type cells attenuated phagocytosis. Hence, we propose that 25HC is essential for optimizing membrane internalization during phagocytosis and that aberrant Ch25h expression in Rorα1-deficient sg/sg macrophages disrupts phagocytosis. Our studies reveal new roles for Rorα1, Ch25h, and 25HC in phagocytosis. Aberrant 25HC underpins the paradoxical association between insulin sensitivity and impaired innate immunity in Rorα1-deficient mice, heralding a wider and essential role for this oxysterol at the nexus of metabolism and immunity.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Hydroxycholesterols/pharmacology
- Immunity, Innate/genetics
- Immunity, Innate/physiology
- Lipopolysaccharides/pharmacology
- Male
- Mice
- Nuclear Receptor Subfamily 1, Group F, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism
- Phagocytosis/genetics
- Phagocytosis/physiology
- Polymerase Chain Reaction
- RNA, Small Interfering/genetics
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Steroid Hydroxylases/genetics
- Steroid Hydroxylases/metabolism
Collapse
Affiliation(s)
- Zewen K Tuong
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Gow DJ, Garceau V, Pridans C, Gow AG, Simpson KE, Gunn-Moore D, Hume DA. Cloning and expression of feline colony stimulating factor receptor (CSF-1R) and analysis of the species specificity of stimulation by colony stimulating factor-1 (CSF-1) and interleukin-34 (IL-34). Cytokine 2012; 61:630-8. [PMID: 23260168 PMCID: PMC3573236 DOI: 10.1016/j.cyto.2012.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 11/22/2012] [Indexed: 01/02/2023]
Abstract
Colony stimulating factor (CSF-1) and its receptor, CSF-1R, have been previously well studied in humans and rodents to dissect the role they play in development of cells of the mononuclear phagocyte system. A second ligand for the CSF-1R, IL-34 has been described in several species. In this study, we have cloned and expressed the feline CSF-1R and examined the responsiveness to CSF-1 and IL-34 from a range of species. The results indicate that pig and human CSF-1 and human IL-34 are equally effective in cats, where both mouse CSF-1 and IL-34 are significantly less active. Recombinant human CSF-1 can be used to generate populations of feline bone marrow and monocyte derived macrophages that can be used to further dissect macrophage-specific gene expression in this species, and to compare it to data derived from mouse, human and pig. These results set the scene for therapeutic use of CSF-1 and IL-34 in cats.
Collapse
|
23
|
Stanley AC, Lieu ZZ, Wall AA, Venturato J, Khromykh T, Hamilton NA, Gleeson PA, Stow JL. Recycling endosome-dependent and -independent mechanisms for IL-10 secretion in LPS-activated macrophages. J Leukoc Biol 2012; 92:1227-39. [PMID: 23012430 DOI: 10.1189/jlb.0412191] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
IL-10 is a key anti-inflammatory cytokine secreted by activated macrophages as a feedback control mechanism to prevent excessive inflammatory responses. Here, we define multiple intracellular trafficking pathways involved in the secretion of newly synthesized IL-10 from macrophages following TLR4 activation with LPS, and show how this relates to the previously defined trafficking pathways for IL-6 and TNF in macrophages simultaneously producing these proinflammatory cytokines. IL-10 exits the Golgi in multiple tubular carriers, including those dependent on p230GRIP. Some of the IL-10 is then delivered to recycling endosomes, where cytokine sorting may occur prior to its release. Another portion of the IL-10 is delivered to the cell surface in distinct vesicles colabeled for apoE. Thus, we show at least two post-Golgi pathways via which IL-10 is trafficked, ensuring its secretion from activated macrophages under different physiological conditions.
Collapse
Affiliation(s)
- A C Stanley
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Sasmono RT, Williams E. Generation and characterization of MacGreen mice, the Cfs1r-EGFP transgenic mice. Methods Mol Biol 2012; 844:157-76. [PMID: 22262441 DOI: 10.1007/978-1-61779-527-5_11] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophage colony-stimulating factor (CSF-1) regulates the differentiation, proliferation, and survival of cells of the mononuclear phagocyte system. The activity of CSF-1 is mediated by the CSF-1 receptor (CSFlR, CD115) that is encoded by c-fms (Csf1r) protooncogene. The c-fms gene is expressed in macrophage, trophoblast cell lineages, and to some extent granulocytes. A reporter gene construct containing 3.5-kb of 5' flanking sequence and the downstream intron 2 of the c-fms gene directed expression of enhanced green fluorescent protein (EGFP) to cells expressing the c-fms gene including the macrophages and trophoblasts. EGFP was detected in trophoblasts from the earliest stage of implantation. During embryonic development, EGFP expression highlighted the large numbers of c-fms positive macrophages in most organs. These embryonic macrophages contribute to organogenesis and tissue remodeling. In adult c-fms EGFP transgenic mice, which have been called the MacGreen mice, EGFP expressed in all tissue macrophage populations and permitted convenient detection of tissue macrophages as well as facilitates their isolation from various tissues.
Collapse
Affiliation(s)
- R Tedjo Sasmono
- Eijkman Institute for Molecular Biology, Jl. Diponegoro 69, Jakarta 10430, Indonesia
| | | |
Collapse
|
25
|
Hamers AAJ, Vos M, Rassam F, Marinković G, Marincovic G, Kurakula K, van Gorp PJ, de Winther MPJ, Gijbels MJJ, de Waard V, de Vries CJM. Bone marrow-specific deficiency of nuclear receptor Nur77 enhances atherosclerosis. Circ Res 2011; 110:428-38. [PMID: 22194623 DOI: 10.1161/circresaha.111.260760] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
RATIONALE Nuclear receptor Nur77, also known as NR4A1, TR3, or NGFI-B, is expressed in human atherosclerotic lesions in macrophages, endothelial cells, T cells and smooth muscle cells. Macrophages play a critical role in atherosclerosis and the function of Nur77 in lesion macrophages has not yet been investigated. OBJECTIVE This study aims to delineate the function of Nur77 in macrophages and to assess the effect of bone marrow-specific deficiency of Nur77 on atherosclerosis. METHODS AND RESULTS We investigated Nur77 in macrophage polarization using bone marrow-derived macrophages (BMM) from wild-type and Nur77-knockout (Nur77(-/-)) mice. Nur77(-/-) BMM exhibit changed expression of M2-specific markers and an inflammatory M1-phenotype with enhanced expression of interleukin-12, IFNγ, and SDF-1α and increased NO synthesis in (non)-stimulated Nur77(-/-) BMM cells. SDF-1α expression in nonstimulated Nur77(-/-) BMM is repressed by Nur77 and the chemoattractive activity of Nur77(-/-) BMM is abolished by SDF-1α inhibiting antibodies. Furthermore, Nur77(-/-) mice show enhanced thioglycollate-elicited migration of macrophages and B cells. The effect of bone marrow-specific deficiency of Nur77 on atherosclerosis was studied in low density lipoprotein receptor-deficient (Ldlr(-/-)) mice. Ldlr(-/-) mice with a Nur77(-/-)-deficient bone marrow transplant developed 2.1-fold larger atherosclerotic lesions than wild-type bone marrow-transplanted mice. These lesions contain more macrophages, T cells, smooth muscle cells and larger necrotic cores. SDF-1α expression is higher in lesions of Nur77(-/-)-transplanted mice, which may explain the observed aggravation of lesion formation. CONCLUSIONS In conclusion, in bone marrow-derived cells the nuclear receptor Nur77 has an anti-inflammatory function, represses SDF-1α expression and inhibits atherosclerosis.
Collapse
Affiliation(s)
- Anouk A J Hamers
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Garcia-Garcia E, Ge JQ, Oladiran A, Montgomery B, El-Din MG, Perez-Estrada LC, Stafford JL, Martin JW, Belosevic M. Ozone treatment ameliorates oil sands process water toxicity to the mammalian immune system. WATER RESEARCH 2011; 45:5849-5857. [PMID: 21940034 DOI: 10.1016/j.watres.2011.08.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 08/15/2011] [Accepted: 08/19/2011] [Indexed: 05/31/2023]
Abstract
We evaluated whether ozonation ameliorated the effects of the organic fraction of oil sands process water (OSPW) on immune functions of mice. Ozonation of OSPW eliminated the capacity of its organic fraction to affect various mouse bone marrow-derived macrophage (BMDM) functions in vitro. These included the production of nitric oxide and the expression of inducible nitric oxide synthase, the production of reactive oxygen intermediates and the expression of NADPH oxidase subunits, phagocytosis, and the expression of pro-inflammatory cytokine genes. Ozone treatment also eliminated the ability of OSPW organic fraction to down-regulate the expression of various pro-inflammatory cytokine and chemokine genes in the liver of mice, one week after oral exposure. We conclude that ozone treatment may be a valuable process for the remediation of large volumes of OSPW.
Collapse
|
27
|
Garcia-Garcia E, Pun J, Perez-Estrada LA, Din MGE, Smith DW, Martin JW, Belosevic M. Commercial naphthenic acids and the organic fraction of oil sands process water downregulate pro-inflammatory gene expression and macrophage antimicrobial responses. Toxicol Lett 2011; 203:62-73. [PMID: 21396992 DOI: 10.1016/j.toxlet.2011.03.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2011] [Revised: 03/02/2011] [Accepted: 03/03/2011] [Indexed: 11/18/2022]
Abstract
This is the first report showing that the organic fraction of oil sands process water (OSPW-OF), and commercial naphthenic acids (C-NAs), cause immunotoxicity. The exposure of mouse bone marrow-derived macrophages (BMDM) to different amounts of C-NAs or OSPW-OF, did not affect cell viability in vitro. We examined whether exposure of BMDM to C-NAs or OSPW-OF affected various antimicrobial responses of these cells. A dose-dependent decrease in nitric oxide response was observed after treatment of BMDM with OSPW-OF, but not with C-NAs. Although OSPW-OF and C-NAs both down-regulated the respiratory burst response of BMDM, the suppression of the production of reactive oxygen intermediates was more pronounced in cells treated with OSPW-OF. Treatment with OSPW-OF or C-NAs reduced BMDM phagocytosis of zymosan and latex beads. The decrease of BMDM antimicrobial response after exposure to OSPW-OF or C-NAs, was accompanied by decreased pro-inflammatory cytokine gene expression. Oral exposure of mice to OSPW-OF caused down-regulation in the expression of genes encoding pro-inflammatory cytokines IFNγ, IL-1β and CSF-1. Our findings indicated that OSPW causes immunotoxic effects that may impair the ability of an exposed host to defend against infectious disease. Furthermore, given the differences between the effects of OSPW-OF and C-NAs, C-NAs should not be assumed to be a direct surrogate for the immunotoxic chemical species in OSPW.
Collapse
Affiliation(s)
- Erick Garcia-Garcia
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
28
|
Low PC, Misaki R, Schroder K, Stanley AC, Sweet MJ, Teasdale RD, Vanhaesebroeck B, Meunier FA, Taguchi T, Stow JL. Phosphoinositide 3-kinase δ regulates membrane fission of Golgi carriers for selective cytokine secretion. ACTA ACUST UNITED AC 2010; 190:1053-65. [PMID: 20837769 PMCID: PMC3101599 DOI: 10.1083/jcb.201001028] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Phosphoinositide 3-kinase (PI3K) p110 isoforms are membrane lipid kinases classically involved in signal transduction. Lipopolysaccharide (LPS)-activated macrophages constitutively and abundantly secrete proinflammatory cytokines including tumor necrosis factor-α (TNF). Loss of function of the p110δ isoform of PI3K using inhibitors, RNA-mediated knockdown, or genetic inactivation in mice abolishes TNF trafficking and secretion, trapping TNF in tubular carriers at the trans-Golgi network (TGN). Kinase-active p110δ localizes to the Golgi complex in LPS-activated macrophages, and TNF is loaded into p230-labeled tubules, which cannot undergo fission when p110δ is inactivated. Similar blocks in fission of these tubules and in TNF secretion result from inhibition of the guanosine triphosphatase dynamin 2. These findings demonstrate a new function for p110δ as part of the membrane fission machinery required at the TGN for the selective trafficking and secretion of cytokines in macrophages.
Collapse
Affiliation(s)
- Pei Ching Low
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland 4072, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
GPR17 is a negative regulator of the cysteinyl leukotriene 1 receptor response to leukotriene D4. Proc Natl Acad Sci U S A 2009; 106:11685-90. [PMID: 19561298 DOI: 10.1073/pnas.0905364106] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The cysteinyl leukotrienes (cys-LTs) are proinflammatory lipid mediators acting on the type 1 cys-LT receptor (CysLT(1)R) to mediate smooth muscle constriction and vascular permeability. GPR17, a G protein-coupled orphan receptor with homology to the P2Y and cys-LT receptors, failed to mediate calcium flux in response to leukotriene (LT) D(4) with stable transfectants. However, in stable cotransfections of 6xHis-tagged GPR17 with Myc-tagged CysLT(1)R, the robust CysLT(1)R-mediated calcium response to LTD(4) was abolished. The membrane expression of the CysLT(1)R analyzed by FACS with anti-Myc Ab was not reduced by the cotransfection, yet both LTD(4)-elicited ERK phosphorylation and the specific binding of [(3)H]LTD(4) to microsomal membranes were fully inhibited. CysLT(1)R and GPR17 expressed in transfected cells were coimmunoprecipitated and identified by Western blots, and confocal immunofluorescence microscopy revealed that GPR17 and CysLT(1)R colocalize on the cell surface of human peripheral blood monocytes. Lentiviral knockdown of GPR17 in mouse bone marrow-derived macrophages (BMMPhis) increased both the membrane expression of CysLT(1)R protein by FACS analysis and the LTD(4)-elicited calcium flux in a dose-dependent manner as compared with control BMMPhis, indicating a negative regulatory function of GPR17 for CysLT(1)R in a primary cell. In IgE-dependent passive cutaneous anaphylaxis, GPR17-deficient mice showed a marked and significant increase in vascular permeability as compared with WT littermates, and this vascular leak was significantly blocked by pretreatment of the mice with the CysLT(1)R antagonist, MK-571. Taken together, our findings suggest that GPR17 is a ligand-independent, constitutive negative regulator for the CysLT(1)R that suppresses CysLT(1)R-mediated function at the cell membrane.
Collapse
|
30
|
Bourgeois C, Majer O, Frohner I, Kuchler K. In vitro systems for studying the interaction of fungal pathogens with primary cells from the mammalian innate immune system. Methods Mol Biol 2009; 470:125-139. [PMID: 19089381 DOI: 10.1007/978-1-59745-204-5_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The incidence of invasive fungal diseases has increased over the past decades, particularly in relation with the increase of immunocompromised patient cohorts (e.g., HIV-infected patients, transplant recipients, immunosuppressed patients with cancer). Opportunistic fungal pathogens such as Candida spp. are most often associated with serious systemic infections. Currently available antifungal drugs are rather unspecific, often with severe side effects. In some cases, their prophylactic use has favored emergence of resistant fungal strains. Major antifungal drugs target the biosynthesis of lipid components of the fungal plasma membrane or the assembly of the cell wall. For a more specific and efficient treatment and prevention of fungal infection, new therapeutic strategies are needed, including strengthening or stimulation of the residual host immune response. Achieving such a goal requires a better understanding of factors important for the defense and the survival of the host combating Candida spp. Where possible, primary cultures of mammalian immune cells of the innate immune system constitute a better suited model than transformed cell lines to study host-pathogen response and virulence. Hence, in vitro primary cell culture systems are a good strategy for a first screening of mutant strains of Candida spp. to identify virulence traits with regard to host cell response and pathogen invasion.
Collapse
Affiliation(s)
- Christelle Bourgeois
- Max F. Perutz Laboratories, Department of Medical Biochemistry, Medical University Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
31
|
Kremen M, Krishnan R, Emery I, Hu JH, Slezicki KI, Wu A, Qian K, Du L, Plawman A, Stempien-Otero A, Dichek DA. Plasminogen mediates the atherogenic effects of macrophage-expressed urokinase and accelerates atherosclerosis in apoE-knockout mice. Proc Natl Acad Sci U S A 2008; 105:17109-14. [PMID: 18957535 PMCID: PMC2579386 DOI: 10.1073/pnas.0808650105] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Indexed: 11/18/2022] Open
Abstract
Urokinase-type plasminogen activator (uPA) is expressed at elevated levels in atherosclerotic human arteries, primarily in macrophages. Plasminogen (Plg), the primary physiologic substrate of uPA, is present at significant levels in blood and interstitial fluid. Both uPA and Plg have activities that could affect atherosclerosis progression. Moreover, correlations between increased Plg activation and accelerated atherosclerosis are reported in several human studies. However, a coherent picture of the role of the uPA/Plg system in atherogenesis has not yet emerged, with at least one animal study suggesting that Plg is atheroprotective. We used a transgenic mouse model of macrophage-targeted uPA overexpression in apolipoprotein E-deficient mice to investigate the roles of uPA and Plg in atherosclerosis. We found that macrophage-expressed uPA accelerated atherosclerotic plaque growth and promoted aortic root dilation through Plg-dependent pathways. These pathways appeared to affect lesion progression rather than initiation and to include actions that disproportionately increase lipid accumulation in the artery wall. In addition, loss of Plg was protective against atherosclerosis both in the presence and absence of uPA overexpression. Transgenic mice with macrophage-targeted uPA overexpression reveal atherogenic roles for both uPA and Plg and are a useful experimental setting for investigating the molecular mechanisms that underlie clinically established relationships between uPA expression, Plg activation, and atherosclerosis progression.
Collapse
Affiliation(s)
- Michal Kremen
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - Ranjini Krishnan
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - Isaac Emery
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - Jie Hong Hu
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - Katherine I. Slezicki
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - Alyssa Wu
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - Kun Qian
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - Liang Du
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - Abigail Plawman
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - April Stempien-Otero
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| | - David A. Dichek
- Department of Medicine, University of Washington, 1959 Northeast Pacific Street, Seattle, WA 98195
| |
Collapse
|
32
|
Frohner IE, Bourgeois C, Yatsyk K, Majer O, Kuchler K. Candida albicans cell surface superoxide dismutases degrade host-derived reactive oxygen species to escape innate immune surveillance. Mol Microbiol 2008; 71:240-52. [PMID: 19019164 PMCID: PMC2713856 DOI: 10.1111/j.1365-2958.2008.06528.x] [Citation(s) in RCA: 198] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Mammalian innate immune cells produce reactive oxygen species (ROS) in the oxidative burst reaction to destroy invading microbial pathogens. Using quantitative real-time ROS assays, we show here that both yeast and filamentous forms of the opportunistic human fungal pathogen Candida albicans trigger ROS production in primary innate immune cells such as macrophages and dendritic cells. Through a reverse genetic approach, we demonstrate that coculture of macrophages or myeloid dendritic cells with C. albicans cells lacking the superoxide dismutase (SOD) Sod5 leads to massive extracellular ROS accumulation in vitro. ROS accumulation was further increased in coculture with fungal cells devoid of both Sod4 and Sod5. Survival experiments show that C. albicans mutants lacking Sod5 and Sod4 exhibit a severe loss of viability in the presence of macrophages in vitro. The reduced viability of sod5Δ/Δ and sod4Δ/Δsod5Δ/Δ mutants relative to wild type is not evident with macrophages from gp91phox−/− mice defective in the oxidative burst activity, demonstrating a ROS-dependent killing activity of macrophages targeting fungal pathogens. These data show a physiological role for cell surface SODs in detoxifying ROS, and suggest a mechanism whereby C. albicans, and perhaps many other microbial pathogens, can evade host immune surveillance in vivo.
Collapse
Affiliation(s)
- Ingrid E Frohner
- Medical University Vienna, Christian Doppler Laboratory for Infection Biology, Max F. Perutz Laboratories, Campus Vienna Biocenter; A-1030 Vienna, Austria
| | | | | | | | | |
Collapse
|
33
|
Helming L, Tomasello E, Kyriakides TR, Martinez FO, Takai T, Gordon S, Vivier E. Essential role of DAP12 signaling in macrophage programming into a fusion-competent state. Sci Signal 2008; 1:ra11. [PMID: 18957693 DOI: 10.1126/scisignal.1159665] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multinucleated giant cells, formed by fusion of macrophages, are a hallmark of granulomatous inflammation. With a genetic approach, we show that signaling through the adaptor protein DAP12 (DNAX activating protein of 12 kD), its associated receptor triggering receptor expressed by myeloid cells 2 (TREM-2), and the downstream protein tyrosine kinase Syk is required for the cytokine-induced formation of giant cells and that overexpression of DAP12 potentiates macrophage fusion. We also present evidence that DAP12 is a general macrophage fusion regulator and is involved in modulating the expression of several macrophage-associated genes, including those encoding known mediators of macrophage fusion, such as DC-STAMP and Cadherin 1. Thus, DAP12 is involved in programming of macrophages through the regulation of gene and protein expression to induce a fusion-competent state.
Collapse
Affiliation(s)
- Laura Helming
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | | | | | | | | | | | |
Collapse
|
34
|
Kornmann LM, Curfs DMJ, Hermeling E, van der Made I, de Winther MPJ, Reneman RS, Reesink KD, Hoeks APG. Perfluorohexane-loaded macrophages as a novel ultrasound contrast agent: a feasibility study. Mol Imaging Biol 2008; 10:264-70. [PMID: 18536974 DOI: 10.1007/s11307-008-0146-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 03/11/2008] [Accepted: 03/26/2008] [Indexed: 01/13/2023]
Abstract
PURPOSE We investigated in vitro the potential of macrophages to act as targeted vehicle for ultrasound molecular imaging. PROCEDURES Murine bone marrow-derived macrophages (BMM), incubated for 3 h with different concentrations of perfluorohexane (PFH) emulsions, were monitored by microscopy, flow cytometry, and ultrasound. Effects of PFH loading on BMM adhesion molecule (PSGL-1, VLA-4, Mac-1, LFA-1) expression were analyzed by flow cytometry. Static adhesion of PFH loaded BMM to unstimulated and TNF-alpha stimulated b.End5 endothelial cells was assessed by microscopy. RESULTS Incubation of BMM with PFH emulsions resulted in dose-dependent uptake and increased echogenicity (max. 17 dB). Flow cytometry analyses revealed no down-regulation related to PFH loading of BMM adhesion molecule expression. Endothelial adhesion remained functional, even after 24 h, although PFH loading dose-dependently attenuated static adhesion. CONCLUSION PFH loaded BMM may potentially serve as ultrasound contrast agent for noninvasive detection of atherogenic hotspots in arteries.
Collapse
Affiliation(s)
- Liselotte M Kornmann
- Department of Biophysics, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, P. O. Box 616, 6200 MD, Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Stempin CC, Garrido VV, Dulgerian LR, Cerbán FM. Cruzipain and SP600125 induce p38 activation, alter NO/arginase balance and favor the survival of Trypanosoma cruzi in macrophages. Acta Trop 2008; 106:119-27. [PMID: 18359001 DOI: 10.1016/j.actatropica.2008.02.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Revised: 02/12/2008] [Accepted: 02/13/2008] [Indexed: 01/19/2023]
Abstract
Cruzipain (Cz), an antigen of Trypanosoma cruzi, mediates the activation of arginase involving p38 MAPK. In this work, it was studied whether the phosphorylation of MAPKs into macrophages (Mvarphi) could be induced by Cz and/or by the parasite. We found that Cz induced activation of p38, while the parasite produced phosphorylation of JNK and p44/p42. MAPK phosphorylation changed and JNK activation was blocked when Mvarphi were pre-incubated with Cz, before coming into contact with T. cruzi. We investigated the role of JNK inhibitor SP600125 on T. cruzi infection, since it also induces p38 phosphorylation. Thus, J774 cells were pre-treated with SP600125 and then infected with T. cruzi. This set of cells showed a decrease in nitric oxide (NO) production and an increase in arginase I expression. Another group of J774 cells was pre-treated with SP600125 and incubated with Cz before being infected with T. cruzi. This second group showed a greater reduction in NO production. These results can be correlated with the parasitic growth since the ex vivo treatment with SP600125 on adherent spleen cells (ASC) of BALB/c infected mice also increased the parasitic growth. Therefore, Cz and SP600125 favor the T. cruzi survival in Mvarphi by changing the iNOS/arginase balance.
Collapse
Affiliation(s)
- Cinthia C Stempin
- CIBICI-CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende s/n, Ciudad Universitaria, 5000 Córdoba, Argentina
| | | | | | | |
Collapse
|
36
|
Schroder K, Spille M, Pilz A, Lattin J, Bode KA, Irvine KM, Burrows AD, Ravasi T, Weighardt H, Stacey KJ, Decker T, Hume DA, Dalpke AH, Sweet MJ. Differential effects of CpG DNA on IFN-beta induction and STAT1 activation in murine macrophages versus dendritic cells: alternatively activated STAT1 negatively regulates TLR signaling in macrophages. THE JOURNAL OF IMMUNOLOGY 2007; 179:3495-503. [PMID: 17785783 DOI: 10.4049/jimmunol.179.6.3495] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Classical STAT1 activation in response to TLR agonists occurs by phosphorylation of the Y701 and S727 residues through autocrine type I IFN signaling and p38 MAPK signaling, respectively. In this study, we report that the TLR9 agonist CpG DNA induced Ifn-beta mRNA, as well as downstream type I IFN-dependent genes, in a MyD88-dependent manner in mouse myeloid dendritic cells. This pathway was required for maximal TNF and IL-6 secretion, as well as expression of cell surface costimulatory molecules. By contrast, neither A- nor B-type CpG-containing oligonucleotides induced Ifn-beta in mouse bone marrow-derived macrophages (BMM) and a CpG-B oligonucleotide did not induce IFn-beta in the macrophage-like cell line, J774. In BMM, STAT1 was alternatively activated (phosphorylated on S727, but not Y701), and was retained in the cytoplasm in response to CpG DNA. CpG DNA responses were altered in BMM from STAT1(S727A) mice; Il-12p40 and Cox-2 mRNAs were more highly induced, whereas Tlr4 and Tlr9 mRNAs were more repressed. The data suggest a novel inhibitory function for cytoplasmic STAT1 in response to TLR agonists that activate p38 MAPK but do not elicit type I IFN production. Indeed, the TLR7 agonist, R837, failed to induce Ifn-beta mRNA and consequently triggered STAT1 phosphorylation on S727, but not Y701, in human monocyte-derived macrophages. The differential activation of Ifn-beta and STAT1 by CpG DNA in mouse macrophages vs dendritic cells provides a likely mechanism for their divergent roles in priming the adaptive immune response.
Collapse
Affiliation(s)
- Kate Schroder
- Cooperative Research Centre for Chronic Inflammatory Diseases and Special Research Centre for Functional and Applied Genomics, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Schroder K, Lichtinger M, Irvine KM, Brion K, Trieu A, Ross IL, Ravasi T, Stacey KJ, Rehli M, Hume DA, Sweet MJ. PU.1 and ICSBP control constitutive and IFN-gamma-regulated Tlr9 gene expression in mouse macrophages. J Leukoc Biol 2007; 81:1577-90. [PMID: 17360957 DOI: 10.1189/jlb.0107036] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Macrophages are activated by unmethylated CpG-containing DNA (CpG DNA) via TLR9. IFN-gamma and LPS can synergize with CpG DNA to enhance proinflammatory responses in murine macrophages. Here, we show that LPS and IFN-gamma up-regulated Tlr9 mRNA in murine bone marrow-derived macrophages (BMM). The ability of LPS and IFN-gamma to induce Tlr9 mRNA expression in BMM was dependent on the presence of the growth factor, CSF-1, which is constitutively present in vivo. However, there were clear differences in mechanisms of Tlr9 mRNA induction. LPS stimulation rapidly removed the CSF-1 receptor (CSF-1R) from the cell surface, thereby blocking CSF-1-mediated transcriptional repression and indirectly inducing Tlr9 mRNA expression. By contrast, IFN-gamma activated the Tlr9 promoter directly and only marginally affected cell surface CSF-1R expression. An approximately 100-bp proximal promoter of the murine Tlr9 gene was sufficient to confer basal and IFN-gamma-inducible expression in RAW264.7 cells. A composite IFN regulatory factor (IRF)/PU.1 site upon the major transcription start site was identified. Mutation of the binding sites for PU.1 or IRF impaired basal promoter activity, but only the IRF-binding site was required for IFN-gamma induction. The mRNA expression of the IRF family member IFN consensus-binding protein [(ICSBP)/IRF8] was coregulated with Tlr9 in macrophages, and constitutive and IFN-gamma-inducible Tlr9 mRNA expression was reduced in ICSBP-deficient BMM. This study therefore characterizes the regulation of mouse Tlr9 expression and defines a molecular mechanism by which IFN-gamma amplifies mouse macrophage responses to CpG DNA.
Collapse
Affiliation(s)
- Kate Schroder
- Special Research Centre for Functional and Applied Genomics, Institute for Molecular Bioscience, University of Queensland, St. Lucia, Brisbane 4072, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Helming L, Gordon S. Macrophage fusion induced by IL-4 alternative activation is a multistage process involving multiple target molecules. Eur J Immunol 2007; 37:33-42. [PMID: 17154265 DOI: 10.1002/eji.200636788] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Multinucleated giant cells, characteristic of granulomatous infections, originate from fusion of macrophages, however, little is known about the underlying mechanism. Alternative activation of macrophages by exposure to IL-4 and IL-13 induces macrophage homokaryon formation. We have established a new quantitative bifluorescent system to study IL-4-induced fusion of primary murine macrophages in vitro. Using this assay, we could show that macrophage fusion is not mediated by a single molecule, but involves multiple functional components. Although several murine macrophage populations were not competent to form giant cells, indicating that they fail to display the full fusion machinery, these non-fusogenic macrophages could fuse with fusion-competent macrophages in a heterophilic manner. Since IL-4 induced molecules were needed on both fusion partners, we conclude that at least two functionally distinct molecules mediate macrophage homokaryon formation with each present on one fusion partner. In addition, though IL-4 treatment led to induction of a fusogenic status, macrophages could only fuse efficiently when adherent to a permissive substratum. Based on our findings, we conclude that macrophage fusion is a multistage process involving multiple target molecules. The model we describe will allow analysis of the molecular basis of membrane fusion and possible insight into alternative activation of macrophages.
Collapse
Affiliation(s)
- Laura Helming
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | | |
Collapse
|
39
|
Groeneweg M, Kanters E, Vergouwe MN, Duerink H, Kraal G, Hofker MH, de Winther MPJ. Lipopolysaccharide-induced gene expression in murine macrophages is enhanced by prior exposure to oxLDL. J Lipid Res 2006; 47:2259-67. [PMID: 16840796 DOI: 10.1194/jlr.m600181-jlr200] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Uptake of modified lipoproteins by macrophages results in the formation of foam cells. We investigated how foam cell formation affects the inflammatory response of macrophages. Murine bone marrow-derived macrophages were treated with oxidized LDL (oxLDL) to induce foam cell formation. Subsequently, the foam cells were activated with lipopolysaccharide (LPS), and the expression of lipid metabolism and inflammatory genes was analyzed. Furthermore, gene expression profiles of foam cells were analyzed using a microarray. We found that prior exposure to oxLDL resulted in enhanced LPS-induced tumor necrosis factor (TNF) and interleukin-6 (IL-6) gene expression, whereas the expression of the anti-inflammatory cytokine IL-10 and interferon-beta was decreased in foam cells. Also, LPS-induced cytokine secretion of TNF, IL-6, and IL-12 was enhanced, whereas secretion of IL-10 was strongly reduced after oxLDL preincubation. Microarray experiments showed that the overall inflammatory response induced by LPS was enhanced by oxLDL loading of the macrophages. Moreover, oxLDL loading was shown to result in increased nuclear factor-kappaB activation. In conclusion, our experiments show that the inflammatory response to LPS is enhanced by loading of macrophages with oxLDL. These data demonstrate that foam cell formation may augment the inflammatory response of macrophages during atherogenesis, possibly in an IL-10-dependent manner.
Collapse
Affiliation(s)
- Mathijs Groeneweg
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
40
|
Stempien-Otero A, Plawman A, Meznarich J, Dyamenahalli T, Otsuka G, Dichek DA. Mechanisms of cardiac fibrosis induced by urokinase plasminogen activator. J Biol Chem 2006; 281:15345-51. [PMID: 16554301 DOI: 10.1074/jbc.m512818200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human hearts with end-stage failure and fibrosis have macrophage accumulation and elevated plasminogen activator activity. However, the mechanisms that link macrophage accumulation and plasminogen activator activity with cardiac fibrosis are unclear. We previously reported that mice with macrophage-targeted overexpression of urokinase plasminogen activator (SR-uPA+/o mice) develop cardiac macrophage accumulation by 5 weeks of age and cardiac fibrosis by 15 weeks. We used SR-uPA+/o mice to investigate mechanisms through which macrophage-expressed uPA causes cardiac macrophage accumulation and fibrosis. We hypothesized that: 1) macrophage accumulation and cardiac fibrosis in SR-uPA+/o mice are dependent on localization of uPA by the uPA receptor (uPAR); 2) activation of plasminogen by uPA and subsequent activation of transforming growth factor-beta1 (TGF-beta1) and matrix metalloproteinase (MMP)-2 and -9 by plasmin are critical pathways through which uPA-expressing macrophages accumulate in the heart and cause fibrosis; and 3) uPA-induced cardiac fibrosis can be attenuated by treatment with verapamil. To test these hypotheses, we bred the SR-uPA+/o transgene into mice deficient in either uPAR or plasminogen and measured cardiac macrophage accumulation and fibrosis. We also measured cardiac TGF-beta1 protein (total and active), Smad2 phosphorylation, and MMP activity after the onset of macrophage accumulation but before the onset of cardiac fibrosis. Finally, we treated mice with verapamil. Our studies revealed that plasminogen is necessary for uPA-induced cardiac fibrosis and macrophage accumulation but uPAR is not. We did not detect plasmin-mediated activation of TGF-beta1, MMP-2, or MMP-9 in hearts of SR-uPA+/o mice. However, verapamil treatment significantly attenuated both cardiac fibrosis and macrophage accumulation.
Collapse
Affiliation(s)
- April Stempien-Otero
- Department of Medicine, Division of Cardiology, University of Washington School of Medicine, Seattle, Washington, 98195, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Fijneman RJA, Vos M, Berkhof J, Demant P, Kraal G. Genetic analysis of macrophage characteristics as a tool to identify tumor susceptibility genes: mapping of three macrophage-associated risk inflammatory factors, marif1, marif2, and marif3. Cancer Res 2004; 64:3458-64. [PMID: 15150098 DOI: 10.1158/0008-5472.can-03-3767] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Genetic predisposition to cancer is influenced by allelic variation in tumor susceptibility genes (TSGs) as present in the germline. We previously demonstrated in the mouse that TSGs frequently participate in genetic interactions, indicating that they represent molecular networks. Inflammation may constitute one of the molecular networks underlying susceptibility to cancer by influencing the tumor microenvironment. Because macrophages play a key role in inflammation and are often associated with tumors, we argue that a subset of TSGs can be identified by examining the genetics of macrophage characteristics. A panel of inflammation-related assays was established to phenotype mouse bone marrow-derived macrophages, which included stimulation with lipopolysaccharides followed by measurement of secretion of tumor necrosis factor alpha and the p40 chain of interleukin-12 and of expression of inducible nitric oxide synthase and cyclooxygenase-2. This panel of assays was used for linkage analysis and applied to bone marrow-derived macrophages derived from individual mice of segregating crosses between inbred strain O20 and the highly related strains NTX-10 and NTX-20, which differed from O20 in only 10% of their genome, to reduce genetic complexity. Three macrophage-associated risk inflammatory factors were mapped-Marif1, Marif2, and Marif3-that each affected several inflammation-related assays, confirming that they function within molecular networks. Moreover, Marif1 and Marif2 were localized in regions with established linkage for both quantitative and qualitative aspects of lung cancer susceptibility. These studies provide a novel approach to investigate the genetics of microenvironmental influence on predisposition to tumorigenesis, thereby contributing to development of new strategies that aim to prevent or treat cancer.
Collapse
Affiliation(s)
- Remond J A Fijneman
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, the Netherlands.
| | | | | | | | | |
Collapse
|
42
|
Truman LA, Ogden CA, Howie SEM, Gregory CD. Macrophage chemotaxis to apoptotic Burkitt's lymphoma cells in vitro: role of CD14 and CD36. Immunobiology 2004; 209:21-30. [PMID: 15481137 DOI: 10.1016/j.imbio.2004.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In Burkitt's lymphoma (BL), apoptosis occurs at high frequency alongside uncontrolled proliferation. Macrophages infiltrate these tumours in large numbers and engage in the phagocytic clearance of apoptotic cells in situ. Here we tested the hypothesis that apoptosis of BL cells may provide a mechanism for recruitment of macrophages to these tumours. We show that monocytes and macrophages, but not neutrophils, preferentially migrated to apoptotic BL cells in vitro. Transfection of BL cells with the anti-apoptotic gene bcl-2 both prevented apoptosis and abolished macrophage chemotaxis. Macrophage migration to BL populations correlated well with the number of apoptotic BL cells present (the Pearson correlation r = 0.81, p<0.0001). Chemoattraction of murine macrophages to apoptotic human BL cells demonstrated that the mechanism was conserved across these species. In an attempt to identify the macrophage receptors involved in this process, we investigated whether CD14 and CD36, two receptors important in the phagocytic clearance of apoptotic cells, were also involved in the chemotactic macrophage response. We found that bone marrow-derived macrophages from CD14-/- and CD36-/- mice moved as well as wild-type macrophages in chemotaxis assays towards apoptotic BL cells. Migrating macrophages were found to be up-regulated in their expression of CD14, however, suggesting that, although this receptor does not appear to be required for 'sensing' apoptotic cells, it may be up-regulated on the surface of the migrating macrophage in readiness for apoptotic corpse clearance.
Collapse
Affiliation(s)
- Lucy A Truman
- lnnate Immunity Group, MRC Centre for Inflammation Research, University of Edinburgh, 2nd floor, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | | | | | | |
Collapse
|
43
|
Nolan KF, Strong V, Soler D, Fairchild PJ, Cobbold SP, Croxton R, Gonzalo JA, Rubio A, Wells M, Waldmann H. IL-10-Conditioned Dendritic Cells, Decommissioned for Recruitment of Adaptive Immunity, Elicit Innate Inflammatory Gene Products in Response to Danger Signals. THE JOURNAL OF IMMUNOLOGY 2004; 172:2201-9. [PMID: 14764687 DOI: 10.4049/jimmunol.172.4.2201] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are the professional APCs of the immune system, enabling T cells to perceive and respond appropriately to potentially dangerous microbes, while also being able to maintain T cell tolerance toward self. In part, such tolerance can be determined by IL-10 released from certain types of regulatory T cells. IL-10 has previously been shown to render DCs unable to activate T cells and it has been assumed that this process represents a general block in maturation. Using serial analysis of gene expression, we show that IL-10 pretreatment of murine bone marrow-derived DCs alone causes significant changes in gene expression. Furthermore, these cells retain the ability to respond to Toll-like receptor agonists, but in a manner skewed toward the selective induction of mediators known to enhance local inflammation and innate immunity, among which we highlight a novel CXCR2 ligand, DC inflammatory protein-1. These data suggest that, while the presence of a protolerogenic and purportedly anti-inflammatory agent such as IL-10 precludes DCs from acquiring their potential as initiators of adaptive immunity, their ability to act as initiators of innate immunity in response to Toll-like receptor signaling is enhanced.
Collapse
MESH Headings
- Amino Acid Motifs
- Animals
- Bone Marrow Cells/immunology
- Bone Marrow Cells/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Line
- Cell Line, Tumor
- Cell Movement/genetics
- Cell Movement/immunology
- Cells, Cultured
- Chemokines, CXC/antagonists & inhibitors
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Chemokines, CXC/physiology
- Chemotaxis, Leukocyte/immunology
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Gene Expression Regulation/immunology
- Gene Library
- Humans
- Immunity, Innate/genetics
- Inflammation Mediators/metabolism
- Interleukin-10/physiology
- Lipopolysaccharides/pharmacology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Molecular Sequence Data
- Monomeric GTP-Binding Proteins/antagonists & inhibitors
- Monomeric GTP-Binding Proteins/biosynthesis
- Monomeric GTP-Binding Proteins/genetics
- Monomeric GTP-Binding Proteins/physiology
- Neutrophil Infiltration/immunology
- Nucleic Acid Amplification Techniques
- RNA, Messenger/biosynthesis
- Receptors, Interleukin-8B/physiology
Collapse
Affiliation(s)
- Kathleen F Nolan
- Sir William Dunn School of Pathology, Oxford University, Oxford, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kanters E, Pasparakis M, Gijbels MJJ, Vergouwe MN, Partouns-Hendriks I, Fijneman RJA, Clausen BE, Förster I, Kockx MM, Rajewsky K, Kraal G, Hofker MH, de Winther MPJ. Inhibition of NF-kappaB activation in macrophages increases atherosclerosis in LDL receptor-deficient mice. J Clin Invest 2003; 112:1176-85. [PMID: 14561702 PMCID: PMC213488 DOI: 10.1172/jci18580] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is now generally accepted as a chronic inflammatory condition. The transcription factor NF-kappaB is a key regulator of inflammation, immune responses, cell survival, and cell proliferation. To investigate the role of NF-kappaB activation in macrophages during atherogenesis, we used LDL receptor-deficient mice with a macrophage-restricted deletion of IkappaB kinase 2 (IKK2), which is essential for NF-kappaB activation by proinflammatory signals. These mice showed increased atherosclerosis as quantified by lesion area measurements. In addition, the lesions were more advanced and showed more necrosis and increased cell number in early lesions. Southern blotting revealed that deletion of IKK2 was approximately 65% in macrophages, coinciding with a reduction of 50% in NF-kappaB activation, as compared with controls. In both groups, the expression of differentiation markers, uptake of bacteria, and endocytosis of modified LDL was similar. Upon stimulation with LPS, production of TNF was reduced by approximately 50% in IKK2-deleted macrophages. Interestingly, we also found a major reduction in the anti-inflammatory cytokine IL-10. Our data show that inhibition of the NF-kappaB pathway in macrophages leads to more severe atherosclerosis in mice, possibly by affecting the pro- and anti-inflammatory balance that controls the development of atherosclerosis.
Collapse
Affiliation(s)
- Edwin Kanters
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kanters E, Pasparakis M, Gijbels MJ, Vergouwe MN, Partouns-Hendriks I, Fijneman RJ, Clausen BE, Förster I, Kockx MM, Rajewsky K, Kraal G, Hofker MH, de Winther MP. Inhibition of NF-κB activation in macrophages increases atherosclerosis in LDL receptor–deficient mice. J Clin Invest 2003. [DOI: 10.1172/jci200318580] [Citation(s) in RCA: 253] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
46
|
Kanters E, Gijbels MJJ, van der Made I, Vergouwe MN, Heeringa P, Kraal G, Hofker MH, de Winther MPJ. Hematopoietic NF-kappaB1 deficiency results in small atherosclerotic lesions with an inflammatory phenotype. Blood 2003; 103:934-40. [PMID: 14512319 DOI: 10.1182/blood-2003-05-1450] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterized by the accumulation of lipid-laden macrophages in the vessel wall. One of the major transcription factors in inflammation is nuclear factor kappaB (NF-kappaB), and we have studied its role in the development of atherosclerosis. Bone marrow from mice targeted in the NF-kappaB1 gene encoding for the p50 subunit was used to reconstitute irradiated LDLR(-/-) mice as a model for atherosclerosis. After feeding the mice a high-fat diet, those deficient in NF-kappaB1 had a 41% lower rate of atherosclerosis than control mice, as judged by the sizes of the lesions. Furthermore, in the absence of NF-kappaB1, the lesions were characterized by an inflammatory phenotype, contained increased numbers of small cells, and were almost devoid of normal foam cells. In vitro studies using bone marrow (BM)-derived macrophages showed that macrophages lacking p50 had a prolonged production of tumor necrosis factor (TNF) in response to lipopolysaccharide (LPS), and other cytokines were also affected. Interestingly, the uptake of oxidized low-density lipoprotein (LDL) was greatly reduced in activated p50-deficient macrophages, probably because of a reduction in the expression of scavenger receptor class A. The effects on atherosclerosis might have resulted from the changes in cytokine production and the uptake of modified lipoproteins, making p50 a pivotal regulator of atherogenesis.
Collapse
Affiliation(s)
- Edwin Kanters
- Department of Molecular Genetics, Cardiovascular Research Institute Maastricht UNS50/11, Universiteitssingel 50, 6229ER Maastricht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Wells CA, Ravasi T, Faulkner GJ, Carninci P, Okazaki Y, Hayashizaki Y, Sweet M, Wainwright BJ, Hume DA. Genetic control of the innate immune response. BMC Immunol 2003; 4:5. [PMID: 12826024 PMCID: PMC194878 DOI: 10.1186/1471-2172-4-5] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2003] [Accepted: 06/26/2003] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Susceptibility to infectious diseases is directed, in part, by the interaction between the invading pathogen and host macrophages. This study examines the influence of genetic background on host-pathogen interactions, by assessing the transcriptional responses of macrophages from five inbred mouse strains to lipopolysaccharide (LPS), a major determinant of responses to gram-negative microorganisms. RESULTS The mouse strains examined varied greatly in the number, amplitude and rate of induction of genes expressed in response to LPS. The response was attenuated in the C3H/HeJlpsd strain, which has a mutation in the LPS receptor Toll-like receptor 4 (TLR4). Variation between mouse strains allowed clustering into early (C57Bl/6J and DBA/2J) and delayed (BALB/c and C3H/ARC) transcriptional phenotypes. There was no clear correlation between gene induction patterns and variation at the Bcg locus (Slc11A1) or propensity to bias Th1 versus Th2 T cell activation responses. CONCLUSION Macrophages from each strain responded to LPS with unique gene expression profiles. The variation apparent between genetic backgrounds provides insights into the breadth of possible inflammatory responses, and paradoxically, this divergence was used to identify a common transcriptional program that responds to TLR4 signalling, irrespective of genetic background. Our data indicates that many additional genetic loci control the nature and the extent of transcriptional responses promoted by a single pathogen-associated molecular pattern (PAMP), such as LPS.
Collapse
Affiliation(s)
- Christine A Wells
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Timothy Ravasi
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Geoffrey J Faulkner
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Piero Carninci
- Laboratory for Genome Exploration Research Group, Genomic Sciences Centre, Riken Yokohama Institute, Yokohama 230-0045
| | - Yasushi Okazaki
- Laboratory for Genome Exploration Research Group, Genomic Sciences Centre, Riken Yokohama Institute, Yokohama 230-0045
| | - Yoshihide Hayashizaki
- Laboratory for Genome Exploration Research Group, Genomic Sciences Centre, Riken Yokohama Institute, Yokohama 230-0045
| | - Matthew Sweet
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Brandon J Wainwright
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - David A Hume
- Institute for Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
48
|
Brown GD, Herre J, Williams DL, Willment JA, Marshall ASJ, Gordon S. Dectin-1 mediates the biological effects of beta-glucans. J Exp Med 2003; 197:1119-24. [PMID: 12719478 PMCID: PMC2193964 DOI: 10.1084/jem.20021890] [Citation(s) in RCA: 899] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The ability of fungal-derived beta-glucan particles to induce leukocyte activation and the production of inflammatory mediators, such as tumor necrosis factor (TNF)-alpha, is a well characterized phenomenon. Although efforts have been made to understand how these carbohydrate polymers exert their immunomodulatory effects, the receptors involved in generating these responses are unknown. Here we show that Dectin-1 mediates the production of TNF-alpha in response to zymosan and live fungal pathogens, an activity that occurs at the cell surface and requires the cytoplasmic tail and immunoreceptor tyrosine activation motif of Dectin-1 as well as Toll-like receptor (TLR)-2 and Myd88. This is the first demonstration that the inflammatory response to pathogens requires recognition by a specific receptor in addition to the TLRs. Furthermore, these studies implicate Dectin-1 in the production of TNF-alpha in response to fungi, a critical step required for the successful control of these pathogens.
Collapse
Affiliation(s)
- Gordon D Brown
- Sir William Dunn School of Pathology, South Parks Road, Oxford OX1 3RE, United Kingdom.
| | | | | | | | | | | |
Collapse
|
49
|
Beers C, Honey K, Fink S, Forbush K, Rudensky A. Differential regulation of cathepsin S and cathepsin L in interferon gamma-treated macrophages. J Exp Med 2003; 197:169-79. [PMID: 12538657 PMCID: PMC2193812 DOI: 10.1084/jem.20020978] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Cathepsin S (catS) and cathepsin L (catL) mediate late stages of invariant chain (Ii) degradation in discrete antigen-presenting cell types. Macrophages (Mphis) are unique in that they express both proteases and here we sought to determine the relative contribution of each enzyme. We observe that catL plays no significant role in Ii cleavage in interferon (IFN)-gamma-stimulated Mphis. In addition, our studies show that the level of catL activity is significantly decreased in Mphis cultured in the presence of IFN-gamma whereas catS activity increases. The decrease in catL activity upon cytokine treatment occurs despite the persistence of high levels of mature catL protein, suggesting that a specific inhibitor of the enzyme is up-regulated in IFN-gamma-stimulated peritoneal Mphis. Similar inhibition of activity is observed in dendritic cells engineered to overexpress catL. Such enzymatic inhibition in Mphis exhibits only partial dependence upon Ii and therefore, other mechanisms of catL inhibition are regulated by IFN-gamma. Thus, during a T helper cell type 1 immune response catL inhibition in Mphis results in preferential usage of catS, such that major histocompatibility complex class II presentation by all bone marrow-derived antigen-presenting cell is regulated by catS.
Collapse
MESH Headings
- Animals
- Antigen Presentation
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Cathepsin L
- Cathepsins/deficiency
- Cathepsins/genetics
- Cathepsins/metabolism
- Cysteine Endopeptidases
- Histocompatibility Antigens Class II/metabolism
- In Vitro Techniques
- Interferon-gamma/pharmacology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/enzymology
- Macrophages, Peritoneal/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Recombinant Proteins
- Th1 Cells/immunology
Collapse
Affiliation(s)
- Courtney Beers
- Department of Immunology, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
50
|
Stacey M, Chang GW, Sanos SL, Chittenden LR, Stubbs L, Gordon S, Lin HH. EMR4, a novel epidermal growth factor (EGF)-TM7 molecule up-regulated in activated mouse macrophages, binds to a putative cellular ligand on B lymphoma cell line A20. J Biol Chem 2002; 277:29283-93. [PMID: 12023293 DOI: 10.1074/jbc.m204306200] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
A novel member of the EGF-TM7 family, mEMR4, was identified and characterized. The full-length mouse EMR4 cDNA encodes a predicted 689-amino acid protein containing two epidermal growth factor (EGF)-like modules, a mucin-like spacer domain, and a seven-transmembrane domain with a cytoplasmic tail. Genetic mapping established that mEMR4 is localized in the distal region of mouse chromosome 17 in close proximity to another EGF-TM7 gene, F4/80 (Emr1). Similar to F4/80, mEMR4 is predominantly expressed on resident macrophages. However, a much lower expression level was also detected in thioglycollate-elicited peritoneal neutrophils and bone marrow-derived dendritic cells. The expression of mEMR4 is up-regulated following macrophage activation in Biogel and thioglycollate-elicited peritoneal macrophages. Similarly, mEMR4 is over-expressed in TNF-alpha-treated resident peritoneal macrophages, whereas interleukin-4 and -10 dramatically reduce the expression. mEMR4 was found to undergo proteolytic processing within the extracellular stalk region resulting in two protein subunits associated noncovalently as a heterodimer. The proteolytic cleavage site was identified by N-terminal amino acid sequencing and located at the conserved GPCR (G protein-coupled receptor) proteolytic site in the extracellular region. Using multivalent biotinylated mEMR4-mFc fusion proteins as a probe, a putative cell surface ligand was identified on a B lymphoma cell line, A20, in a cell-binding assay. The mEMR4-ligand interaction is Ca2+-independent and is mediated predominantly by the second EGF-like module. mEMR4 is the first EGF-TM7 receptor known to mediate the cellular interaction between myeloid cells and B cells.
Collapse
Affiliation(s)
- Martin Stacey
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, United Kingdom
| | | | | | | | | | | | | |
Collapse
|