1
|
Kunanopparat A, Dinh TTH, Ponpakdee P, Padungros P, Kaewduangduen W, Ariya-anandech K, Tummamunkong P, Samaeng A, Sae-ear P, Leelahavanichkul A, Hirankarn N, Ritprajak P. Complement receptor 3-dependent engagement by Candida glabrata β-glucan modulates dendritic cells to induce regulatory T-cell expansion. Open Biol 2024; 14:230315. [PMID: 38806144 PMCID: PMC11293457 DOI: 10.1098/rsob.230315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/05/2024] [Accepted: 04/15/2024] [Indexed: 05/30/2024] Open
Abstract
Candida glabrata is an important pathogen causing invasive infection associated with a high mortality rate. One mechanism that causes the failure of Candida eradication is an increase in regulatory T cells (Treg), which play a major role in immune suppression and promoting Candida pathogenicity. To date, how C. glabrata induces a Treg response remains unclear. Dendritic cells (DCs) recognition of fungi provides the fundamental signal determining the fate of the T-cell response. This study investigated the interplay between C. glabrata and DCs and its effect on Treg induction. We found that C. glabrata β-glucan was a major component that interacted with DCs and consequently mediated the Treg response. Blocking the binding of C. glabrata β-glucan to dectin-1 and complement receptor 3 (CR3) showed that CR3 activation in DCs was crucial for the induction of Treg. Furthermore, a ligand-receptor binding assay showed the preferential binding of C. glabrata β-glucan to CR3. Our data suggest that C. glabrata β-glucan potentially mediates the Treg response, probably through CR3-dependent activation in DCs. This study contributes new insights into immune modulation by C. glabrata that may lead to a better design of novel immunotherapeutic strategies for invasive C. glabrata infection.
Collapse
Affiliation(s)
- Areerat Kunanopparat
- Department of Microbiology, Faculty of Dentistry, Center of Excellence in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Truc Thi Huong Dinh
- Department of Microbiology, Faculty of Dentistry, Center of Excellence in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Chulalongkorn University, Bangkok, Thailand
- Medical Microbiology Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Department of Pathophysiology and Immunology, Faculty of Medicine, Can Tho University of Medicine and Pharmacy, Vietnam
| | - Pranpariya Ponpakdee
- Department of Chemistry, Faculty of Science, Green Chemistry for Fine Chemical Production and Environmental Remediation Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Panuwat Padungros
- Department of Chemistry, Faculty of Science, Green Chemistry for Fine Chemical Production and Environmental Remediation Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Warerat Kaewduangduen
- Department of Microbiology, Faculty of Dentistry, Center of Excellence in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Chulalongkorn University, Bangkok, Thailand
| | - Kasirapat Ariya-anandech
- Department of Microbiology, Faculty of Dentistry, Center of Excellence in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Chulalongkorn University, Bangkok, Thailand
| | - Phawida Tummamunkong
- Department of Microbiology, Faculty of Dentistry, Center of Excellence in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Chulalongkorn University, Bangkok, Thailand
| | - Amanee Samaeng
- Department of Microbiology, Faculty of Dentistry, Center of Excellence in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Chulalongkorn University, Bangkok, Thailand
| | - Pannagorn Sae-ear
- Faculty of Dentistry, Oral Biology Research Center, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Chulalongkorn University, Bangkok, Thailand
| | - Nattiya Hirankarn
- Center of Excellence in Immunology and Immune-Mediated Diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Patcharee Ritprajak
- Department of Microbiology, Faculty of Dentistry, Center of Excellence in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
2
|
Schittenhelm L, Robertson J, Pratt AG, Hilkens CM, Morrison VL. Dendritic cell integrin expression patterns regulate inflammation in the rheumatoid arthritis joint. Rheumatology (Oxford) 2021; 60:1533-1542. [PMID: 33123735 PMCID: PMC7937020 DOI: 10.1093/rheumatology/keaa686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 09/19/2020] [Indexed: 11/14/2022] Open
Abstract
Objectives Immune dysregulation contributes to the development of RA. Altered surface expression patterns of integrin adhesion receptors by immune cells is one mechanism by which this may occur. We investigated the role of β2 integrin subunits CD11a and CD11b in dendritic cell (DC) subsets of RA patients. Methods Total β2 integrin subunit expression and its conformation (‘active’ vs ‘inactive’ state) were quantified in DC subsets from peripheral blood (PB) and SF of RA patients as well as PB from healthy controls. Ex vivo stimulation of PB DC subsets and in vitro-generated mature and tolerogenic monocyte-derived DCs (moDCs) were utilized to model the clinical findings. Integrin subunit contribution to DC function was tested by analysing clustering and adhesion, and in co-cultures to assess T cell activation. Results A significant reduction in total and active CD11a expression in DCs in RA SF compared with PB and, conversely, a significant increase in CD11b expression was found. These findings were modelled in vitro using moDCs: tolerogenic moDCs showed higher expression of active CD11a and reduced levels of active CD11b compared with mature moDCs. Finally, blockade of CD11b impaired T cell activation in DC–T cell co-cultures. Conclusion For the first time in RA, we show opposing expression of CD11a and CD11b in DCs in environments of inflammation (CD11alow/CD11bhigh) and steady state/tolerance (CD11ahigh/CD11blow), as well as a T cell stimulatory role for CD11b. These findings highlight DC integrins as potential novel targets for intervention in RA.
Collapse
Affiliation(s)
- Leonie Schittenhelm
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK.,Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK
| | - Jamie Robertson
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Arthur G Pratt
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK.,Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Catharien M Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK
| | - Vicky L Morrison
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis (RACE), Glasgow, UK
| |
Collapse
|
3
|
Bednarczyk M, Medina-Montano C, Fittler FJ, Stege H, Roskamp M, Kuske M, Langer C, Vahldieck M, Montermann E, Tubbe I, Röhrig N, Dzionek A, Grabbe S, Bros M. Complement-Opsonized Nano-Carriers Are Bound by Dendritic Cells (DC) via Complement Receptor (CR)3, and by B Cell Subpopulations via CR-1/2, and Affect the Activation of DC and B-1 Cells. Int J Mol Sci 2021; 22:2869. [PMID: 33799879 PMCID: PMC8001596 DOI: 10.3390/ijms22062869] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 02/07/2023] Open
Abstract
The development of nanocarriers (NC) for biomedical applications has gained large interest due to their potential to co-deliver drugs in a cell-type-targeting manner. However, depending on their surface characteristics, NC accumulate serum factors, termed protein corona, which may affect their cellular binding. We have previously shown that NC coated with carbohydrates to enable biocompatibility triggered the lectin-dependent complement pathway, resulting in enhanced binding to B cells via complement receptor (CR)1/2. Here we show that such NC also engaged all types of splenic leukocytes known to express CR3 at a high rate when NC were pre-incubated with native mouse serum resulting in complement opsonization. By focusing on dendritic cells (DC) as an important antigen-presenting cell type, we show that CR3 was essential for binding/uptake of complement-opsonized NC, whereas CR4, which in mouse is specifically expressed by DC, played no role. Further, a minor B cell subpopulation (B-1), which is important for first-line pathogen responses, and co-expressed CR1/2 and CR3, in general, engaged NC to a much higher extent than normal B cells. Here, we identified CR-1/2 as necessary for binding of complement-opsonized NC, whereas CR3 was dispensable. Interestingly, the binding of complement-opsonized NC to both DC and B-1 cells affected the expression of activation markers. Our findings may have important implications for the design of nano-vaccines against infectious diseases, which codeliver pathogen-specific protein antigen and adjuvant, aimed to induce a broad adaptive cellular and humoral immune response by inducing cytotoxic T lymphocytes that kill infected cells and pathogen-neutralizing antibodies, respectively. Decoration of nano-vaccines either with carbohydrates to trigger complement activation in vivo or with active complement may result in concomitant targeting of DC and B cells and thereby may strongly enhance the extent of dual cellular/humoral immune responses.
Collapse
Affiliation(s)
- Monika Bednarczyk
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Carolina Medina-Montano
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Frederic Julien Fittler
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Meike Roskamp
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Michael Kuske
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Christian Langer
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Marco Vahldieck
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Evelyn Montermann
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Ingrid Tubbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Nadine Röhrig
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Andrzej Dzionek
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429 Bergisch Gladbach, Germany; (M.R.); (C.L.); (M.V.); (A.D.)
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany; (M.B.); (C.M.-M.); (F.J.F.); (H.S.); (M.K.); (E.M.); (I.T.); (N.R.); (S.G.)
| |
Collapse
|
4
|
Human Peripheral Blood Dendritic Cell and T Cell Activation by Codium fragile Polysaccharide. Mar Drugs 2020; 18:md18110535. [PMID: 33120897 PMCID: PMC7692045 DOI: 10.3390/md18110535] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/11/2020] [Accepted: 10/23/2020] [Indexed: 12/11/2022] Open
Abstract
Natural polysaccharides exhibit an immunostimulatory effect with low toxicity in humans and animals. It has shown that polysaccharide extracted from Codium fragile (CFP) induces anti-cancer immunity by dendritic cell (DC) activation, while the effect of CFP has not examined in the human immune cells. In this study, we found that CFP promoted the upregulation of CD80, CD83 and CD86 and major histocompatibility complex (MHC) class I and II in human monocyte-derived dendritic cells (MDDCs). In addition, CFP induced the production of proinflammatory cytokines in MDDCs. Moreover, CFP directly induced the activation of Blood Dendritic Cell Antigen (BDCA)1+ and BDCA3+ subsets of human peripheral blood DCs (PBDCs). The CFP-stimulated BDCA1+ PBDCs further promoted activation and proliferation of syngeneic CD4 T cells. The CFP-activated BDCA3+ PBDCs activated syngeneic CD8 T cells, which produced cytotoxic mediators, namely, cytotoxic T lymphocytes. These results suggest that CFP may be a candidate molecule for enhancing immune activation in humans.
Collapse
|
5
|
Mellors J, Tipton T, Longet S, Carroll M. Viral Evasion of the Complement System and Its Importance for Vaccines and Therapeutics. Front Immunol 2020; 11:1450. [PMID: 32733480 PMCID: PMC7363932 DOI: 10.3389/fimmu.2020.01450] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/04/2020] [Indexed: 12/17/2022] Open
Abstract
The complement system is a key component of innate immunity which readily responds to invading microorganisms. Activation of the complement system typically occurs via three main pathways and can induce various antimicrobial effects, including: neutralization of pathogens, regulation of inflammatory responses, promotion of chemotaxis, and enhancement of the adaptive immune response. These can be vital host responses to protect against acute, chronic, and recurrent viral infections. Consequently, many viruses (including dengue virus, West Nile virus and Nipah virus) have evolved mechanisms for evasion or dysregulation of the complement system to enhance viral infectivity and even exacerbate disease symptoms. The complement system has multifaceted roles in both innate and adaptive immunity, with both intracellular and extracellular functions, that can be relevant to all stages of viral infection. A better understanding of this virus-host interplay and its contribution to pathogenesis has previously led to: the identification of genetic factors which influence viral infection and disease outcome, the development of novel antivirals, and the production of safer, more effective vaccines. This review will discuss the antiviral effects of the complement system against numerous viruses, the mechanisms employed by these viruses to then evade or manipulate this system, and how these interactions have informed vaccine/therapeutic development. Where relevant, conflicting findings and current research gaps are highlighted to aid future developments in virology and immunology, with potential applications to the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Jack Mellors
- Public Health England, National Infection Service, Salisbury, United Kingdom.,Department of Infection Biology, Institute of Infection and Global Health, University of Liverpool, Liverpool, United Kingdom
| | - Tom Tipton
- Public Health England, National Infection Service, Salisbury, United Kingdom
| | - Stephanie Longet
- Public Health England, National Infection Service, Salisbury, United Kingdom
| | - Miles Carroll
- Public Health England, National Infection Service, Salisbury, United Kingdom
| |
Collapse
|
6
|
Park HB, Lim SM, Hwang J, Zhang W, You S, Jin JO. Cancer immunotherapy using a polysaccharide from Codium fragile in a murine model. Oncoimmunology 2020; 9:1772663. [PMID: 32923129 PMCID: PMC7458636 DOI: 10.1080/2162402x.2020.1772663] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Natural polysaccharides have shown immune modulatory effects with low toxicity in both animal and human models. A previous study has shown that the polysaccharide from Codium fragile (CFP) promotes natural killer (NK) cell activation in mice. Since NK cell activation is mediated by dendritic cells (DCs), we examined the effect of CFP on DC activation and evaluated the subsequent induction of anti-cancer immunity in a murine model. Treatment with CFP induced activation of bone marrow-derived dendritic cells (BMDCs). Moreover, subcutaneous injection of CFP promoted the activation of spleen and lymph node DCs in vivo. CFP also induced activation of DCs in tumor-bearing mice, and combination treatment with CFP and ovalbumin (OVA) promoted OVA-specific T cell activation, which consequently promoted infiltration of IFN-γ-and TNF-α-producing OT-1 and OT-II cells into the tumors. Moreover, combination treatment using CFP and cancer self-antigen efficiently inhibited B16 tumor growth in the mouse model. Treatment with CFP also enhanced anti-PD-L1 antibody mediated anti-cancer immunity in the CT-26 carcinoma-bearing BALB/c mice. Taken together these data suggest that CFP may function as an adjuvant in the treatment of cancer by enhancing immune activation. Abbreviations CFP: Codium fragile polysaccharide; NK: natural killer; IFN: interferon; TNF: tumor necrosis factor; IL: interleukin; tdLN: tumor draining lymph node; BMDC: bone marrow-derived dendritic cell; OVA: ovalbumin; Ab: antibody; Ag: antigen; DC: dendritic cell; CTL: cytotoxic T lymphocyte; APC: antigen-presenting cell; pDC: plasmacytoid dendritic cell; mDC: myeloid dendritic cell; MHC: major histocompatibility complex; CR3: complement receptor type 3; TLR: Toll-like receptor; LPS: lipopolysaccharide; SP: sulfated polysaccharide; TRP2: tyrosinase-related protein 2; SR-A: scavenger receptor-A
Collapse
Affiliation(s)
- Hae-Bin Park
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Seong-Min Lim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Juyoung Hwang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Wei Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - SangGuan You
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, South Korea
| | - Jun-O Jin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
7
|
Halder LD, Jo EAH, Hasan MZ, Ferreira-Gomes M, Krüger T, Westermann M, Palme DI, Rambach G, Beyersdorf N, Speth C, Jacobsen ID, Kniemeyer O, Jungnickel B, Zipfel PF, Skerka C. Immune modulation by complement receptor 3-dependent human monocyte TGF-β1-transporting vesicles. Nat Commun 2020; 11:2331. [PMID: 32393780 PMCID: PMC7214408 DOI: 10.1038/s41467-020-16241-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles have an important function in cellular communication. Here, we show that human and mouse monocytes release TGF-β1-transporting vesicles in response to the pathogenic fungus Candida albicans. Soluble β-glucan from C. albicans binds to complement receptor 3 (CR3, also known as CD11b/CD18) on monocytes and induces the release of TGF-β1-transporting vesicles. CR3-dependence is demonstrated using CR3-deficient (CD11b knockout) monocytes generated by CRISPR-CAS9 genome editing and isolated from CR3-deficient (CD11b knockout) mice. These vesicles reduce the pro-inflammatory response in human M1-macrophages as well as in whole blood. Binding of the vesicle-transported TGF-β1 to the TGF-β receptor inhibits IL1B transcription via the SMAD7 pathway in whole blood and induces TGFB1 transcription in endothelial cells, which is resolved upon TGF-β1 inhibition. Notably, human complement-opsonized apoptotic bodies induce production of similar TGF-β1-transporting vesicles in monocytes, suggesting that the early immune response might be suppressed through this CR3-dependent anti-inflammatory vesicle pathway.
Collapse
Affiliation(s)
- Luke D Halder
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Emeraldo A H Jo
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Mohammad Z Hasan
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Marta Ferreira-Gomes
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Friedrich Schiller University, 07745, Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Martin Westermann
- Electron Microscopy Center, University Hospital Jena, 07743, Jena, Germany
| | - Diana I Palme
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Günter Rambach
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, 97070, Würzburg, Germany
| | - Cornelia Speth
- Division of Hygiene and Medical Microbiology, Medical University of Innsbruck, A-6020, Innsbruck, Austria
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.,Friedrich Schiller University, 07743, Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany
| | - Berit Jungnickel
- Department of Cell Biology, Institute of Biochemistry and Biophysics, Friedrich Schiller University, 07745, Jena, Germany
| | - Peter F Zipfel
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.,Friedrich Schiller University, 07743, Jena, Germany
| | - Christine Skerka
- Department of Infection Biology, Leibniz Institute for Natural Product Research and Infection Biology, 07745, Jena, Germany.
| |
Collapse
|
8
|
Lo MW, Woodruff TM. Complement: Bridging the innate and adaptive immune systems in sterile inflammation. J Leukoc Biol 2020; 108:339-351. [PMID: 32182389 DOI: 10.1002/jlb.3mir0220-270r] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/07/2020] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The complement system is a collection of soluble and membrane-bound proteins that together act as a powerful amplifier of the innate and adaptive immune systems. Although its role in infection is well established, complement is becoming increasingly recognized as a key contributor to sterile inflammation, a chronic inflammatory process often associated with noncommunicable diseases. In this context, damaged tissues release danger signals and trigger complement, which acts on a range of leukocytes to augment and bridge the innate and adaptive immune systems. Given the detrimental effect of chronic inflammation, the complement system is therefore well placed as an anti-inflammatory drug target. In this review, we provide a general outline of the sterile activators, effectors, and targets of the complement system and a series of examples (i.e., hypertension, cancer, allograft transplant rejection, and neuroinflammation) that highlight complement's ability to bridge the 2 arms of the immune system.
Collapse
Affiliation(s)
- Martin W Lo
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Brisbane, Queensland, Australia
| |
Collapse
|
9
|
Mice expressing the variant rs1143679 allele of ITGAM (CD11b) show impaired DC-mediated T cell proliferation. Mamm Genome 2019; 30:245-259. [PMID: 31673770 PMCID: PMC6842653 DOI: 10.1007/s00335-019-09819-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 10/03/2019] [Indexed: 11/09/2022]
Abstract
Genome-wide association studies (GWAS) and functional genomic analyses have implicated several ITGAM (CD11b) single-nucleotide polymorphisms (SNPs) in the development of SLE and other disorders. ITGAM encodes the αM chain of the β2 integrin Mac-1, a receptor that plays important roles in myeloid cell functions. The ITGAM SNP rs1143679, which results in an arginine to histidine change at amino acid position 77 of the CD11b protein, has been shown to reduce binding to several ligands and to alter Mac-1-mediated cellular response in vitro. Importantly, however, the potential contribution of this SNP variant to the initiation and/or progression of immune and inflammatory processes in vivo remains unexplored. Herein, we describe for the first time the generation and characterization of a mouse line expressing the 77His variant of CD11b. Surprisingly, we found that 77His did not significantly affect Mac-1-mediated leukocyte migration and activation as assessed using thioglycollate-induced peritonitis and LPS/TNF-α-induced dermal inflammation models. In contrast, expression of this variant did alter T cell immunity, as evidenced by significantly reduced proliferation of ovalbumin (OVA)-specific transgenic T cells in 77His mice immunized with OVA. Reduced antigen-specific T cell proliferation was also observed when either 77His splenic dendritic cells (DCs) or bone marrow-derived DCs were used as antigen-presenting cells (APCs). Although more work is necessary to determine how this alteration might influence the development of SLE or other diseases, these in vivo findings suggest that the 77His variant of CD11b can compromise the ability of DCs to induce antigen-driven T cell proliferation.
Collapse
|
10
|
Ellegård R, Khalid M, Svanberg C, Holgersson H, Thorén Y, Wittgren MK, Hinkula J, Nyström S, Shankar EM, Larsson M. Complement-Opsonized HIV-1 Alters Cross Talk Between Dendritic Cells and Natural Killer (NK) Cells to Inhibit NK Killing and to Upregulate PD-1, CXCR3, and CCR4 on T Cells. Front Immunol 2018; 9:899. [PMID: 29760706 PMCID: PMC5936988 DOI: 10.3389/fimmu.2018.00899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/11/2018] [Indexed: 01/07/2023] Open
Abstract
Dendritic cells (DCs), natural killer (NK) cells, and T cells play critical roles during primary HIV-1 exposure at the mucosa, where the viral particles become coated with complement fragments and mucosa-associated antibodies. The microenvironment together with subsequent interactions between these cells and HIV at the mucosal site of infection will determine the quality of immune response that ensues adaptive activation. Here, we investigated how complement and immunoglobulin opsonization influences the responses triggered in DCs and NK cells, how this affects their cross talk, and what T cell phenotypes are induced to expand following the interaction. Our results showed that DCs exposed to complement-opsonized HIV (C-HIV) were less mature and had a poor ability to trigger IFN-driven NK cell activation. In addition, when the DCs were exposed to C-HIV, the cytotolytic potentials of both NK cells and CD8 T cells were markedly suppressed. The expression of PD-1 as well as co-expression of negative immune checkpoints TIM-3 and LAG-3 on PD-1 positive cells were increased on both CD4 as well as CD8 T cells upon interaction with and priming by NK–DC cross talk cultures exposed to C-HIV. In addition, stimulation by NK–DC cross talk cultures exposed to C-HIV led to the upregulation of CD38, CXCR3, and CCR4 on T cells. Together, the immune modulation induced during the presence of complement on viral surfaces is likely to favor HIV establishment, dissemination, and viral pathogenesis.
Collapse
Affiliation(s)
- Rada Ellegård
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mohammad Khalid
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Pharmaceutics, College of Pharmacy, King Khalid University, Asir-Abha, Saudi Arabia
| | - Cecilia Svanberg
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Hanna Holgersson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Ylva Thorén
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mirja Karolina Wittgren
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Jorma Hinkula
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Sofia Nyström
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Clinical Immunology and Transfusion Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Esaki M Shankar
- Division of Infection Biology, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India.,Center of Excellence for Research in AIDS (CERiA), University of Malaya, Lembah Pantai, Kuala Lumpur, Malaysia.,Department of Microbiology, Central University of Tamil Nadu, Thiruvarur, India
| | - Marie Larsson
- Division of Molecular Virology, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
11
|
Hoang KV, Rajaram MVS, Curry HM, Gavrilin MA, Wewers MD, Schlesinger LS. Complement Receptor 3-Mediated Inhibition of Inflammasome Priming by Ras GTPase-Activating Protein During Francisella tularensis Phagocytosis by Human Mononuclear Phagocytes. Front Immunol 2018; 9:561. [PMID: 29632532 PMCID: PMC5879101 DOI: 10.3389/fimmu.2018.00561] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/06/2018] [Indexed: 01/08/2023] Open
Abstract
Francisella tularensis is a remarkably infectious facultative intracellular bacterium of macrophages that causes tularemia. Early evasion of host immune responses contributes to the success of F. tularensis as a pathogen. F. tularensis entry into human monocytes and macrophages is mediated by the major phagocytic receptor, complement receptor 3 (CR3, CD11b/CD18). We recently determined that despite a significant increase in macrophage uptake following C3 opsonization of the virulent Type A F. tularensis spp. tularensis Schu S4, this phagocytic pathway results in limited pro-inflammatory cytokine production. Notably, MAP kinase/ERK activation is suppressed immediately during C3-opsonized Schu S4-CR3 phagocytosis. A mathematical model of CR3-TLR2 crosstalk predicted early involvement of Ras GTPase-activating protein (RasGAP) in immune suppression by CR3. Here, we link CR3-mediated uptake of opsonized Schu S4 by human monocytes and macrophages with inhibition of early signal 1 inflammasome activation, evidenced by limited caspase-1 cleavage and IL-18 release. This inhibition is due to increased RasGAP activity, leading to a reduction in the Ras-ERK signaling cascade upstream of the early inflammasome activation event. Thus, our data uncover a novel signaling pathway mediated by CR3 following engagement of opsonized virulent F. tularensis to limit inflammasome activation in human phagocytic cells, thereby contributing to evasion of the host innate immune system.
Collapse
Affiliation(s)
- Ky V Hoang
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Murugesan V S Rajaram
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Heather Marie Curry
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Mikhail A Gavrilin
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Mark D Wewers
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Internal Medicine, Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH, United States
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Texas Biomedical Research Institute, San Antonio, TX, United States
| |
Collapse
|
12
|
Schetters STT, Gomez-Nicola D, Garcia-Vallejo JJ, Van Kooyk Y. Neuroinflammation: Microglia and T Cells Get Ready to Tango. Front Immunol 2018; 8:1905. [PMID: 29422891 PMCID: PMC5788906 DOI: 10.3389/fimmu.2017.01905] [Citation(s) in RCA: 231] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022] Open
Abstract
In recent years, many paradigms concerning central nervous system (CNS) immunology have been challenged and shifted, including the discovery of CNS-draining lymphatic vessels, the origin and functional diversity of microglia, the impact of T cells on CNS immunological homeostasis and the role of neuroinflammation in neurodegenerative diseases. In parallel, antigen presentation outside the CNS has revealed the vital role of antigen-presenting cells in maintaining tolerance toward self-proteins, thwarting auto-immunity. Here, we review recent findings that unite these shifted paradigms of microglial functioning, antigen presentation, and CNS-directed T cell activation, focusing on common neurodegenerative diseases. It provides an important update on CNS adaptive immunity, novel targets, and a concept of the microglia T-cell equilibrium.
Collapse
Affiliation(s)
- Sjoerd T T Schetters
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Diego Gomez-Nicola
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| | - Yvette Van Kooyk
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
13
|
Schittenhelm L, Hilkens CM, Morrison VL. β 2 Integrins As Regulators of Dendritic Cell, Monocyte, and Macrophage Function. Front Immunol 2017; 8:1866. [PMID: 29326724 PMCID: PMC5742326 DOI: 10.3389/fimmu.2017.01866] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/08/2017] [Indexed: 12/24/2022] Open
Abstract
Emerging evidence suggests that the β2 integrin family of adhesion molecules have an important role in suppressing immune activation and inflammation. β2 integrins are important adhesion and signaling molecules that are exclusively expressed on leukocytes. The four β2 integrins (CD11a, CD11b, CD11c, and CD11d paired with the β2 chain CD18) play important roles in regulating three key aspects of immune cell function: recruitment to sites of inflammation; cell-cell contact formation; and downstream effects on cellular signaling. Through these three processes, β2 integrins both contribute to and regulate immune responses. This review explores the pro- and anti-inflammatory effects of β2 integrins in monocytes, macrophages, and dendritic cells and how they influence the outcome of immune responses. We furthermore discuss how imbalances in β2 integrin function can have far-reaching effects on mounting appropriate immune responses, potentially influencing the development and progression of autoimmune and inflammatory diseases. Therapeutic targeting of β2 integrins, therefore, holds enormous potential in exploring treatment options for a variety of inflammatory conditions.
Collapse
Affiliation(s)
- Leonie Schittenhelm
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom.,Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, United Kingdom
| | - Catharien M Hilkens
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, United Kingdom
| | - Vicky L Morrison
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, United Kingdom.,Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE), Glasgow, United Kingdom
| |
Collapse
|
14
|
Leonard F, Ha NP, Sule P, Alexander JF, Volk DE, Lokesh GLR, Liu X, Cirillo JD, Gorenstein DG, Yuan J, Chatterjee S, Graviss EA, Godin B. Thioaptamer targeted discoidal microparticles increase self immunity and reduce Mycobacterium tuberculosis burden in mice. J Control Release 2017; 266:238-247. [PMID: 28987879 DOI: 10.1016/j.jconrel.2017.09.038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/29/2017] [Indexed: 12/25/2022]
Abstract
Worldwide, tuberculosis (TB) remains one of the most prevalent infectious diseases causing morbidity and death in >1.5 million patients annually. Mycobacterium tuberculosis (Mtb), the etiologic agent of TB, usually resides in the alveolar macrophages. Current tuberculosis treatment methods require more than six months, and low compliance often leads to therapeutic failure and multidrug resistant strain development. Critical to improving TB-therapy is shortening treatment duration and increasing therapeutic efficacy. In this study, we sought to determine if lung hemodynamics and pathological changes in Mtb infected cells can be used for the selective targeting of microparticles to infected tissue(s). Thioaptamers (TA) with CD44 (CD44TA) targeting moiety were conjugated to discoidal silicon mesoporous microparticles (SMP) to enhance accumulation of these agents/carriers in the infected macrophages in the lungs. In vitro, CD44TA-SMP accumulated in macrophages infected with mycobacteria efficiently killing the infected cells and decreasing survival of mycobacteria. In vivo, increased accumulations of CD44TA-SMP were recorded in the lung of M. tuberculosis infected mice as compared to controls. TA-targeted carriers significantly diminished bacterial load in the lungs and caused recruitment of T lymphocytes. Proposed mechanism of action of the designed vector accounts for a combination of increased uptake of particles that leads to infected macrophage death, as well as, activation of cellular immunity by the TA, causing increased T-cell accumulation in the treated lungs. Based on our data with CD44TA-SMP, we anticipate that this drug carrier can open new avenues in TB management.
Collapse
Affiliation(s)
- Fransisca Leonard
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - Ngan P Ha
- Department of Pathology and Genomic Medicine Houston, Houston Methodist Research Institute, TX 77030, United States
| | - Preeti Sule
- Texas A&M Health Science Center, Department of Microbial Pathogenesis and Immunology, Bryan, TX 77807, United States
| | - Jenolyn F Alexander
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - David E Volk
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Ganesh L R Lokesh
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Xuewu Liu
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States
| | - Jeffrey D Cirillo
- Texas A&M Health Science Center, Department of Microbial Pathogenesis and Immunology, Bryan, TX 77807, United States
| | - David G Gorenstein
- University of Texas Health Science Center at Houston, Department of NanoMedicine and Biomedical Engineering, Institute of Molecular Medicine, Houston, TX 77030, United States
| | - Jinyun Yuan
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, St. Louis, MO 63104, United States
| | - Soumya Chatterjee
- Saint Louis University School of Medicine, Department of Internal Medicine, Division of Infectious Diseases, Allergy and Immunology, St. Louis, MO 63104, United States
| | - Edward A Graviss
- Department of Pathology and Genomic Medicine Houston, Houston Methodist Research Institute, TX 77030, United States
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, TX 77030, United States.
| |
Collapse
|
15
|
Stevanin M, Busso N, Chobaz V, Pigni M, Ghassem-Zadeh S, Zhang L, Acha-Orbea H, Ehirchiou D. CD11b regulates the Treg/Th17 balance in murine arthritis via IL-6. Eur J Immunol 2017; 47:637-645. [PMID: 28191643 DOI: 10.1002/eji.201646565] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 01/07/2017] [Accepted: 02/09/2017] [Indexed: 12/28/2022]
Abstract
Th17 cells are often associated with autoimmunity and been shown to be increased in CD11b-/- mice. Here, we examined the role of CD11b in murine collagen-induced arthritis (CIA). C57BL/6 and CD11b-/- resistant mice were immunized with type II collagen. CD11b-/- mice developed arthritis with early onset, high incidence, and sustained severity compared with C57BL/6 mice. We observed a marked leukocyte infiltration, and histological examinations of the arthritic paws from CD11b-/- mice revealed that the cartilage was destroyed in association with strong lymphocytic infiltration. The CD11b deficiency led to enhanced Th17-cell differentiation. CD11b-/- dendritic cells (DCs) induced much stronger IL-6 production and hence Th17-cell differentiation than wild-type DCs. Treatment of CD11b-/- mice after establishment of the Treg/Th17 balance with an anti-IL-6 receptor mAb significantly suppressed the induction of Th17 cells and reduced arthritis severity. Finally, the severe phenotype of arthritis in CD11b-/- mice was rescued by adoptive transfer of CD11b+ DCs. Taken together, our results indicate that the resistance to CIA in C57BL/6 mice is regulated by CD11b via suppression of IL-6 production leading to reduced Th17-cell differentiation. Therefore, CD11b may represent a susceptibility factor for autoimmunity and could be a target for future therapy.
Collapse
Affiliation(s)
- Mathias Stevanin
- Department of Biochemistry CIIL, University of Lausanne, Epalinges, Switzerland
| | - Nathalie Busso
- DAL, Service of Rheumatology, Laboratory of Rheumatology, University of Lausanne, CHUV, Epalinges, Switzerland
| | - Véronique Chobaz
- DAL, Service of Rheumatology, Laboratory of Rheumatology, University of Lausanne, CHUV, Epalinges, Switzerland
| | - Matteo Pigni
- Department of Biochemistry CIIL, University of Lausanne, Epalinges, Switzerland
| | - Sahar Ghassem-Zadeh
- Department of Biochemistry CIIL, University of Lausanne, Epalinges, Switzerland
| | - Li Zhang
- Center for Vascular and Inflammatory Diseases, Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Hans Acha-Orbea
- Department of Biochemistry CIIL, University of Lausanne, Epalinges, Switzerland
| | - Driss Ehirchiou
- Department of Biochemistry CIIL, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
16
|
Abstract
Mac-1 (CD11b/CD18) is a β2 integrin classically regarded as a pro-inflammatory molecule because of its ability to promote phagocyte cytotoxic functions and enhance the function of several effector molecules such as FcγR, uPAR, and CD14. Nevertheless, recent reports have revealed that Mac-1 also plays significant immunoregulatory roles, and genetic variants in ITGAM, the gene that encodes CD11b, confer risk for the autoimmune disease systemic lupus erythematosus (SLE). This has renewed interest in the physiological roles of this integrin and raised new questions on how its seemingly opposing biological functions may be regulated. Here, we provide an overview of the CD18 integrins and how their activation may be regulated as this may shed light on how the opposing roles of Mac-1 may be elicited. We then discuss studies that exemplify Mac-1's pro-inflammatory versus regulatory roles particularly in the context of IgG immune complex-mediated inflammation. This includes a detailed examination of molecular mechanisms that could explain the risk-conferring effect of rs1143679, a single nucleotide non-synonymous Mac-1 polymorphism associated with SLE.
Collapse
Affiliation(s)
- Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Tanya N Mayadas
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Neutrophil-Mediated Regulation of Innate and Adaptive Immunity: The Role of Myeloperoxidase. J Immunol Res 2016; 2016:2349817. [PMID: 26904693 PMCID: PMC4745373 DOI: 10.1155/2016/2349817] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 12/21/2015] [Indexed: 01/15/2023] Open
Abstract
Neutrophils are no longer seen as leukocytes with a sole function of being the essential first responders in the removal of pathogens at sites of infection. Being armed with numerous pro- and anti-inflammatory mediators, these phagocytes can also contribute to the development of various autoimmune diseases and can positively or negatively regulate the generation of adaptive immune responses. In this review, we will discuss how myeloperoxidase, the most abundant neutrophil granule protein, plays a key role in the various functions of neutrophils in innate and adaptive immunity.
Collapse
|
18
|
Cao C, Zhao J, Doughty EK, Migliorini M, Strickland DK, Kann MG, Zhang L. Mac-1 Regulates IL-13 Activity in Macrophages by Directly Interacting with IL-13Rα1. J Biol Chem 2015; 290:21642-51. [PMID: 26160172 DOI: 10.1074/jbc.m115.645796] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Indexed: 11/06/2022] Open
Abstract
Mac-1 exhibits a unique inhibitory activity toward IL-13-induced JAK/STAT activation and thereby regulates macrophage to foam cell transformation. However, the underlying molecular mechanism is unknown. In this study, we report the identification of IL-13Rα1, a component of the IL-13 receptor (IL-13R), as a novel ligand of integrin Mac-1, using a co-evolution-based algorithm. Biochemical analyses demonstrated that recombinant IL-13Rα1 binds Mac-1 in a purified system and supports Mac-1-mediated cell adhesion. Co-immunoprecipitation experiments revealed that endogenous Mac-1 forms a complex with IL-13Rα1 in solution, and confocal fluorescence microscopy demonstrated that these two receptors co-localize with each other on the surface of macrophages. Moreover, we found that genetic inactivation of Mac-1 promotes IL-13-induced JAK/STAT activation in macrophages, resulting in enhanced polarization along the alternative activation pathway. Importantly, we observed that Mac-1(-/-) macrophages exhibit increased expression of foam cell differentiation markers including 15-lipoxygenase and lectin-type oxidized LDL receptor-1 both in vitro and in vivo. Indeed, we found that Mac-1(-/-)LDLR(-/-) mice develop significantly more foam cells than control LDLR(-/-) mice, using an in vivo model of foam cell formation. Together, our data establish for the first time a molecular mechanism by which Mac-1 regulates the signaling activity of IL-13 in macrophages. This newly identified IL-13Rα1/Mac-1-dependent pathway may offer novel targets for therapeutic intervention in the future.
Collapse
Affiliation(s)
| | | | - Emily K Doughty
- the Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland 21250
| | - Mary Migliorini
- Surgery, Center for Vascular and Inflammatory Diseases, the University of Maryland, School of Medicine, Baltimore, Maryland 21201 and
| | - Dudley K Strickland
- Surgery, Center for Vascular and Inflammatory Diseases, the University of Maryland, School of Medicine, Baltimore, Maryland 21201 and
| | - Maricel G Kann
- the Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, Maryland 21250
| | - Li Zhang
- From the Departments of Physiology and
| |
Collapse
|
19
|
Scott D, Botto M. The paradoxical roles of C1q and C3 in autoimmunity. Immunobiology 2015; 221:719-25. [PMID: 26001732 DOI: 10.1016/j.imbio.2015.05.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/21/2015] [Accepted: 05/01/2015] [Indexed: 01/29/2023]
Abstract
In this review we will focus on the links between complement and autoimmune diseases and will highlight how animal models have provided insights into the manner by which C1q and C3 act to modulate both adaptive and innate immune responses. In particular we will highlight how C1q may not only act as initiator of the classical complement pathway, but can also mediate multiple immune responses in a complement activation independent manner.
Collapse
Affiliation(s)
- Diane Scott
- Centre for Complement and Inflammation Research, Department of Medicine, Imperial College London, London, UK
| | - Marina Botto
- Centre for Complement and Inflammation Research, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
20
|
Hopp AK, Rupp A, Lukacs-Kornek V. Self-antigen presentation by dendritic cells in autoimmunity. Front Immunol 2014; 5:55. [PMID: 24592266 PMCID: PMC3923158 DOI: 10.3389/fimmu.2014.00055] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Accepted: 01/30/2014] [Indexed: 11/13/2022] Open
Abstract
The operation of both central and peripheral tolerance ensures the prevention of autoimmune diseases. The maintenance of peripheral tolerance requires self-antigen presentation by professional antigen presenting cells (APCs). Dendritic cells (DCs) are considered as major APCs involved in this process. The current review discusses the role of DCs in autoimmune diseases, the various factors involved in the induction and maintenance of tolerogenic DC phenotype, and pinpoints their therapeutic capacity as well as potential novel targets for future clinical studies.
Collapse
Affiliation(s)
- Ann-Katrin Hopp
- Department of Medicine II, Saarland University Medical Center , Homburg , Germany
| | - Anne Rupp
- Department of Medicine II, Saarland University Medical Center , Homburg , Germany
| | | |
Collapse
|
21
|
Sriram U, Xu J, Chain RW, Varghese L, Chakhtoura M, Bennett HL, Zoltick PW, Gallucci S. IL-4 suppresses the responses to TLR7 and TLR9 stimulation and increases the permissiveness to retroviral infection of murine conventional dendritic cells. PLoS One 2014; 9:e87668. [PMID: 24489947 PMCID: PMC3906189 DOI: 10.1371/journal.pone.0087668] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 12/28/2013] [Indexed: 12/11/2022] Open
Abstract
Th2-inducing pathological conditions such as parasitic diseases increase susceptibility to viral infections through yet unclear mechanisms. We have previously reported that IL-4, a pivotal Th2 cytokine, suppresses the response of murine bone-marrow-derived conventional dendritic cells (cDCs) and splenic DCs to Type I interferons (IFNs). Here, we analyzed cDC responses to TLR7 and TLR9 ligands, R848 and CpGs, respectively. We found that IL-4 suppressed the gene expression of IFNβ and IFN-responsive genes (IRGs) upon TLR7 and TLR9 stimulation. IL-4 also inhibited IFN-dependent MHC Class I expression and amplification of IFN signaling pathways triggered upon TLR stimulation, as indicated by the suppression of IRF7 and STAT2. Moreover, IL-4 suppressed TLR7- and TLR9-induced cDC production of pro-inflammatory cytokines such as TNFα, IL-12p70 and IL-6 by inhibiting IFN-dependent and NFκB-dependent responses. IL-4 similarly suppressed TLR responses in splenic DCs. IL-4 inhibition of IRGs and pro-inflammatory cytokine production upon TLR7 and TLR9 stimulation was STAT6-dependent, since DCs from STAT6-KO mice were resistant to the IL-4 suppression. Analysis of SOCS molecules (SOCS1, −2 and −3) showed that IL-4 induces SOCS1 and SOCS2 in a STAT6 dependent manner and suggest that IL-4 suppression could be mediated by SOCS molecules, in particular SOCS2. IL-4 also decreased the IFN response and increased permissiveness to viral infection of cDCs exposed to a HIV-based lentivirus. Our results indicate that IL-4 modulates and counteracts pro-inflammatory stimulation induced by TLR7 and TLR9 and it may negatively affect responses against viruses and intracellular parasites.
Collapse
Affiliation(s)
- Uma Sriram
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple University, School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (US); (SG)
| | - Jun Xu
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple University, School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Robert W. Chain
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple University, School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Linda Varghese
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple University, School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Marita Chakhtoura
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple University, School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Heather L. Bennett
- Joseph Stokes, Jr. Research Institute, Division of Rheumatology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Philip W. Zoltick
- Department of Surgery, The Children's Center for Fetal Research, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Stefania Gallucci
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple University, School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail: (US); (SG)
| |
Collapse
|
22
|
Polysaccharides from Ganoderma formosanum function as a Th1 adjuvant and stimulate cytotoxic T cell response in vivo. Vaccine 2014; 32:401-8. [DOI: 10.1016/j.vaccine.2013.11.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 09/20/2013] [Accepted: 11/06/2013] [Indexed: 12/21/2022]
|
23
|
Cis-acting pathways selectively enforce the non-immunogenicity of shed placental antigen for maternal CD8 T cells. PLoS One 2013; 8:e84064. [PMID: 24391885 PMCID: PMC3877187 DOI: 10.1371/journal.pone.0084064] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/12/2013] [Indexed: 11/19/2022] Open
Abstract
Maternal immune tolerance towards the fetus and placenta is thought to be established in part by pathways that attenuate T cell priming to antigens released from the placenta into maternal blood. These pathways remain largely undefined and their existence, at face value, seems incompatible with a mother's need to maintain a functional immune system during pregnancy. A particular conundrum is evident if we consider that maternal antigen presenting cells, activated in order to prime T cells to pathogen-derived antigens, would also have the capacity to prime T cells to co-ingested placental antigens. Here, we address this paradox using a transgenic system in which placental membranes are tagged with a strong surrogate antigen (ovalbumin). We find that although a remarkably large quantity of acellular ovalbumin-containing placental material is released into maternal blood, splenic CD8 T cells in pregnant mice bearing unmanipulated T cell repertoires are not primed to ovalbumin even if the mice are intravenously injected with adjuvants. This failure was largely independent of regulatory T cells, and instead was linked to the intrinsic characteristics of the released material that rendered it selectively non-immunogenic, potentially by sequestering it from CD8α+ dendritic cells. The release of ovalbumin-containing placental material into maternal blood thus had no discernable impact on CD8 T cell priming to soluble ovalbumin injected intravenously during pregnancy, nor did it induce long-term tolerance to ovalbumin. Together, these results outline a major pathway governing the maternal immune response to the placenta, and suggest how tolerance to placental antigens can be maintained systemically without being detrimental to host defense.
Collapse
|
24
|
Gallo PM, Gallucci S. The dendritic cell response to classic, emerging, and homeostatic danger signals. Implications for autoimmunity. Front Immunol 2013; 4:138. [PMID: 23772226 PMCID: PMC3677085 DOI: 10.3389/fimmu.2013.00138] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 05/23/2013] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) initiate and control immune responses, participate in the maintenance of immunological tolerance and are pivotal players in the pathogenesis of autoimmunity. In patients with autoimmune disease and in experimental animal models of autoimmunity, DCs show abnormalities in both numbers and activation state, expressing immunogenic levels of costimulatory molecules and pro-inflammatory cytokines. Exogenous and endogenous danger signals activate DCs to stimulate the immune response. Classic endogenous danger signals are released, activated, or secreted by host cells and tissues experiencing stress, damage, and non-physiologic cell death; and are therefore referred to as damage-associated molecular patterns (DAMPs). Some DAMPs are released from cells, where they are normally sequestered, during necrosis (e.g., heat shock proteins, uric acid, ATP, HMGB1, mitochondria-derived molecules). Others are actively secreted, like Type I Interferons. Here we discuss important DAMPs in the context of autoimmunity. For some, there is a clear pathogenic link (e.g., nucleic acids and lupus). For others, there is less evidence. Additionally, we explore emerging danger signals. These include inorganic materials and man-made technologies (e.g., nanomaterials) developed as novel therapeutic approaches. Some nanomaterials can activate DCs and may trigger unintended inflammatory responses. Finally, we will review “homeostatic danger signals,” danger signals that do not derive directly from pathogens or dying cells but are associated with perturbations of tissue/cell homeostasis and may signal pathological stress. These signals, like acidosis, hypoxia, and changes in osmolarity, also play a role in inflammation and autoimmunity.
Collapse
Affiliation(s)
- Paul M Gallo
- Laboratory of Dendritic Cell Biology, Department of Microbiology and Immunology, Temple Autoimmunity Center, Temple University School of Medicine , Philadelphia, PA , USA
| | | |
Collapse
|
25
|
Ocaña-Morgner C, Götz A, Wahren C, Jessberger R. SWAP-70 restricts spontaneous maturation of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2013; 190:5545-58. [PMID: 23636062 DOI: 10.4049/jimmunol.1203095] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Spontaneous maturation observed in dendritic cell (DC) cultures has been linked to their capacity to induce immune responses. Despite several recent studies, the mechanisms and signals triggering spontaneous maturation of DCs are largely unknown. We found that the absence of SWAP-70 causes spontaneous maturation of spleen- and bone marrow-derived DCs and, in vivo, of spleen-resident CD11c(+)CD11b(+)CD8α(-) DCs. Activation markers, cross-presentation of exogenous Ags, and activation of CD8(+) T cells are much increased in Swap-70(-/-) DCs. Spontaneous maturation of Swap-70(-/-) DCs depends on cell-cell contact and does not involve β-catenin signaling. SWAP-70 is known to regulate integrin activity. Signaling through the integrin CD11b (αM) subunit increases spontaneous maturation of wild-type (wt), but not of Swap-70(-/-) DCs. Signaling through the CD18 (β2) subunit decreases spontaneous maturation of wt and Swap-70(-/-) DCs. Constitutive activation of RhoA in Swap-70(-/-) DCs was determined as a key mechanism causing the increased spontaneous maturation. Inhibition of RhoA early, but not late, in the activation process reduces spontaneous maturation in Swap-70(-/-) DCs to wt levels. Inhibition of RhoA activation during CD11b integrin activation had a significant effect only in Swap-70(-/-) but not in wt DCs. Together, our data suggest that integrin-mediated spontaneous maturation of wt DCs does not depend on active RhoA, whereas the increase in spontaneous maturation of Swap-70(-/-) DCs is supported by integrin CD11b and by hyperactive RhoA. Thus, SWAP-70 deficiency reveals two pathways that contribute to spontaneous maturation of DCs.
Collapse
Affiliation(s)
- Carlos Ocaña-Morgner
- Institute of Physiological Chemistry, Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, D-01307 Dresden, Germany.
| | | | | | | |
Collapse
|
26
|
Neutrophil myeloperoxidase regulates T-cell-driven tissue inflammation in mice by inhibiting dendritic cell function. Blood 2013; 121:4195-204. [PMID: 23509155 DOI: 10.1182/blood-2012-09-456483] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myeloperoxidase (MPO) is important in intracellular microbial killing by neutrophils but extracellularly causes tissue damage. Its role in adaptive immunity and T-cell-mediated diseases is poorly understood. Here, T-cell responses in lymph nodes (LNs) were enhanced by MPO deletion or in vivo inhibition, causing enhanced skin delayed-type hypersensitivity and antigen (Ag)-induced arthritis. Responses of adoptively transferred OT-II T cells were greater in MPO-deficient than wild-type (WT) recipients. MPO, deposited by neutrophils in LNs after Ag injection, interacted with dendritic cells (DCs) in vivo. Culture of murine or human DCs with purified MPO or neutrophil supernatant showed that enzymatically dependent MPO-mediated inhibition of DC activation occurs via MPO-generated reactive intermediates and involves DC Mac-1. Transfer of DCs cultured with WT, but not MPO-deficient, neutrophil supernatant attenuated Ag-specific immunity in vivo. MPO deficiency or in vivo inhibition increased DC activation in LNs after immunization. Studies with DQ-ovalbumin showed that MPO inhibits Ag uptake/processing by DCs. In vivo DC transfer and in vitro studies showed that MPO inhibits DC migration to LNs by reducing their expression of CCR7. Therefore, MPO, via its catalytic activity, inhibits the generation of adaptive immunity by suppressing DC activation, Ag uptake/processing, and migration to LNs to limit pathological tissue inflammation.
Collapse
|
27
|
Phagocytosis is the main CR3-mediated function affected by the lupus-associated variant of CD11b in human myeloid cells. PLoS One 2013; 8:e57082. [PMID: 23451151 PMCID: PMC3579793 DOI: 10.1371/journal.pone.0057082] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 01/17/2013] [Indexed: 11/19/2022] Open
Abstract
The CD11b/CD18 integrin (complement receptor 3, CR3) is a surface receptor on monocytes, neutrophils, macrophages and dendritic cells that plays a crucial role in several immunological processes including leukocyte extravasation and phagocytosis. The minor allele of a non-synonymous CR3 polymorphism (rs1143679, conversation of arginine to histidine at position 77: R77H) represents one of the strongest genetic risk factor in human systemic lupus erythematosus, with heterozygosity (77R/H) being the most common disease associated genotype. Homozygosity for the 77H allele has been reported to reduce adhesion and phagocytosis in human monocytes and monocyte-derived macrophages, respectively, without affecting surface expression of CD11b. Herein we comprehensively assessed the influence of R77H on different CR3-mediated activities in monocytes, neutrophils, macrophages and dendritic cells. R77H did not alter surface expression of CD11b including its active form in any of these cell types. Using two different iC3b-coated targets we found that the uptake by heterozygous 77R/H macrophages, monocytes and neutrophils was significantly reduced compared to 77R/R cells. Allele-specific transduced immortalized macrophage cell lines demonstrated that the minor allele, 77H, was responsible for the impaired phagocytosis. R77H did not affect neutrophil adhesion, neutrophil transmigration in vivo or Toll-like receptor 7/8-mediated cytokine release by monocytes or dendritic cells with or without CR3 pre-engagement by iC3b-coated targets. Our findings demonstrate that the reduction in CR3-mediated phagocytosis associated with the 77H CD11b variant is not macrophage-restricted but demonstrable in other CR3-expressing professional phagocytic cells. The association between 77H and susceptibility to systemic lupus erythematosus most likely relates to impaired waste disposal, a key component of lupus pathogenesis.
Collapse
|
28
|
Fine tuning inflammation at the front door: macrophage complement receptor 3-mediates phagocytosis and immune suppression for Francisella tularensis. PLoS Pathog 2013; 9:e1003114. [PMID: 23359218 PMCID: PMC3554622 DOI: 10.1371/journal.ppat.1003114] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 11/19/2012] [Indexed: 12/11/2022] Open
Abstract
Complement receptor 3 (CR3, CD11b/CD18) is a major macrophage phagocytic receptor. The biochemical pathways through which CR3 regulates immunologic responses have not been fully characterized. Francisella tularensis is a remarkably infectious, facultative intracellular pathogen of macrophages that causes tularemia. Early evasion of the host immune response contributes to the virulence of F. tularensis and CR3 is an important receptor for its phagocytosis. Here we confirm that efficient attachment and uptake of the highly virulent Type A F. tularensis spp. tularensis strain Schu S4 by human monocyte-derived macrophages (hMDMs) requires complement C3 opsonization and CR3. However, despite a>40-fold increase in uptake following C3 opsonization, Schu S4 induces limited pro-inflammatory cytokine production compared with non-opsonized Schu S4 and the low virulent F. novicida. This suggests that engagement of CR3 by opsonized Schu S4 contributes specifically to the immune suppression during and shortly following phagocytosis which we demonstrate by CD11b siRNA knockdown in hMDMs. This immune suppression is concomitant with early inhibition of ERK1/2, p38 MAPK and NF-κB activation. Furthermore, TLR2 siRNA knockdown shows that pro-inflammatory cytokine production and MAPK activation in response to non-opsonized Schu S4 depends on TLR2 signaling providing evidence that CR3-TLR2 crosstalk mediates immune suppression for opsonized Schu S4. Deletion of the CD11b cytoplasmic tail reverses the CR3-mediated decrease in ERK and p38 activation during opsonized Schu-S4 infection. The CR3-mediated signaling pathway involved in this immune suppression includes Lyn kinase and Akt activation, and increased MKP-1, which limits TLR2-mediated pro-inflammatory responses. These data indicate that while the highly virulent F. tularensis uses CR3 for efficient uptake, optimal engagement of this receptor down-regulates TLR2-dependent pro-inflammatory responses by inhibiting MAPK activation through outside-in signaling. CR3-linked immune suppression is an important mechanism involved in the pathogenesis of F. tularensis infection.
Collapse
|
29
|
CR3 is the dominant phagocytotic complement receptor on human dendritic cells. Immunobiology 2012; 218:652-63. [PMID: 22906751 DOI: 10.1016/j.imbio.2012.07.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 07/31/2012] [Accepted: 07/31/2012] [Indexed: 11/22/2022]
Abstract
Dendritic cells (DCs) play a decisive role in immunity; they interact with various pathogens via several pattern recognition and different opsonophagocytotic receptors, including Fc- and complement-receptors. β2-integrins, including complement receptors CR3 (CD11b/CD18) and CR4 (CD11c/CD18) participate in many immunological processes, especially those involving cell migration, adherence, and phagocytosis. Human monocyte derived dendritic cells (MDCs) are known to express CR3 as well as CR4, however possible differences regarding the role of these receptors has not been addressed so far. Our aim was to explore whether there is a difference between the binding and uptake of various complement-opsonized microorganisms, mediated by CR3 and CR4. Studying the expression of receptors during differentiation of MDCs we found that the appearance of CD11b decreased, whereas that of CD11c increased. Interestingly, both receptors were present in the cell membrane in an active conformation. Here we demonstrate that ligation of CD11b directs MDCs to enhanced phagocytosis, while the maturation of the cells and their inflammatory cytokine production are not affected. Blocking CD11c alone did not change the uptake of opsonized yeast or bacteria by MDCs. We confirmed these results using siRNA; namely downregulation of CD11b blocked the phagocytosis of microbes while silencing CD11c had no effect on their uptake. Our data clearly demonstrate that complement C3-dependent phagocytosis of MDCs is mediated mainly by CR3.
Collapse
|
30
|
Driss V, Hermann E, Legrand F, Loiseau S, Delbeke M, Kremer L, Guerardel Y, Dombrowicz D, Capron M. CR3-dependent negative regulation of human eosinophils by Mycobacterium bovis BCG lipoarabinomannan. Immunol Lett 2012; 143:202-7. [DOI: 10.1016/j.imlet.2012.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 02/16/2012] [Accepted: 02/16/2012] [Indexed: 01/21/2023]
|
31
|
Newton K, Dixit VM. Signaling in innate immunity and inflammation. Cold Spring Harb Perspect Biol 2012; 76:442-6. [PMID: 22296764 DOI: 10.1016/j.humimm.2015.03.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2014] [Revised: 02/05/2015] [Accepted: 03/11/2015] [Indexed: 12/24/2022]
Abstract
Inflammation is triggered when innate immune cells detect infection or tissue injury. Surveillance mechanisms involve pattern recognition receptors (PRRs) on the cell surface and in the cytoplasm. Most PRRs respond to pathogen-associated molecular patterns (PAMPs) or host-derived damage-associated molecular patterns (DAMPs) by triggering activation of NF-κB, AP1, CREB, c/EBP, and IRF transcription factors. Induction of genes encoding enzymes, chemokines, cytokines, adhesion molecules, and regulators of the extracellular matrix promotes the recruitment and activation of leukocytes, which are critical for eliminating foreign particles and host debris. A subset of PRRs activates the protease caspase-1, which causes maturation of the cytokines IL1β and IL18. Cell adhesion molecules and chemokines facilitate leukocyte extravasation from the circulation to the affected site, the chemokines stimulating G-protein-coupled receptors (GPCRs). Binding initiates signals that regulate leukocyte motility and effector functions. Other triggers of inflammation include allergens, which form antibody complexes that stimulate Fc receptors on mast cells. Although the role of inflammation is to resolve infection and injury, increasing evidence indicates that chronic inflammation is a risk factor for cancer.
Collapse
Affiliation(s)
- Kim Newton
- Department of Physiological Chemistry, Genentech, Inc., South San Francisco, California 94080, USA
| | | |
Collapse
|
32
|
Municio C, Hugo E, Alvarez Y, Alonso S, Blanco L, Fernández N, Sánchez Crespo M. Apoptotic cells enhance IL-10 and reduce IL-23 production in human dendritic cells treated with zymosan. Mol Immunol 2011; 49:97-106. [DOI: 10.1016/j.molimm.2011.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 07/27/2011] [Accepted: 07/28/2011] [Indexed: 12/22/2022]
|
33
|
Turnis ME, Rooney CM. Enhancement of dendritic cells as vaccines for cancer. Immunotherapy 2011; 2:847-62. [PMID: 21091116 DOI: 10.2217/imt.10.56] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Dendritic cells are the most potent antigen-presenting cells known; owing to their ability to stimulate antigen-specific cytolytic and memory T-cell responses, their use as cancer vaccines is rapidly increasing. While clinical trials provide evidence that dendritic cells vaccines are safe and elicit immunological responses in most patients, few complete tumor remissions have been reported and further technological advances are required. An effective dendritic cell vaccine must possess and maintain several characteristics: it must migrate to lymph nodes, have a mature, Th1-polarizing phenotype expressed stably after infusion and present antigen for sufficient time to produce a T-cell response capable of eliminating a tumor. While dendritic cells are readily matured ex vivo, their phenotype and fate after infusion are rarely evaluable; therefore, strategies to ensure that dendritic cells access lymphoid tissues and retain an immunostimulatory phenotype are required. In order to best exploit dendritic cells as vaccines, they may require genetic modification and combination with other strategies including adoptive T-cell transfer, inhibition of regulatory T cells or modulation of inflammatory pathways.
Collapse
|
34
|
McCloskey ML, Curotto de Lafaille MA, Carroll MC, Erlebacher A. Acquisition and presentation of follicular dendritic cell-bound antigen by lymph node-resident dendritic cells. ACTA ACUST UNITED AC 2010; 208:135-48. [PMID: 21173103 PMCID: PMC3023135 DOI: 10.1084/jem.20100354] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Follicular dendritic cells (DCs [FDCs]) are prominent stromal cell constituents of B cell follicles with the remarkable ability to retain complement-fixed antigens on their cell surface for extended periods of time. These retained immune complexes have long been known to provide the antigenic stimulus that drives antibody affinity maturation, but their role in cellular immunity has remained unclear. In this study, we show that FDC-retained antigens are continually sampled by lymph node-resident DCs for presentation to CD8 T cells. This novel pathway of antigen acquisition was detectable when FDCs were loaded with purified antigens bound into classical antigen-antibody immune complexes, as well as after pregnancy, when they are loaded physiologically with antigens associated with the complement-fixed microparticles released from the placenta into maternal blood. In both cases, ensuing antigen presentation was profoundly tolerogenic, as it induced T cell deletion even under inflammatory conditions. These results significantly broaden the scope of FDC function and suggest new ways that the complement system and persistent antigen presentation might influence T cell activation and the maintenance of peripheral immune tolerance.
Collapse
Affiliation(s)
- Megan L McCloskey
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | |
Collapse
|
35
|
Integrin CD11b negatively regulates TLR-triggered inflammatory responses by activating Syk and promoting degradation of MyD88 and TRIF via Cbl-b. Nat Immunol 2010; 11:734-42. [PMID: 20639876 DOI: 10.1038/ni.1908] [Citation(s) in RCA: 364] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 06/16/2010] [Indexed: 02/06/2023]
Abstract
Integrins are critical for the migration and function of leukocytes in inflammation. However, the interaction between integrin alpha(M) (CD11b), which has high expression in monocytes and macrophages, and Toll-like receptor (TLR)-triggered innate immunity remains unclear. Here we report that CD11b deficiency enhanced TLR-mediated responses in macrophages, rendering mice more susceptible to endotoxin shock and Escherichia coli-caused sepsis. CD11b was activated by TLR-triggered phosphatidylinositol 3-OH kinase (PI(3)K) and the effector RapL and fed back to inhibit TLR signaling by activating the tyrosine kinases Src and Syk. Syk interacted with and induced tyrosine phosphorylation of MyD88 and TRIF, which led to degradation of these adaptor molecules by the E3 ubiquitin ligase Cbl-b. Thus, TLR-triggered, active CD11b integrin engages in crosstalk with the MyD88 and TRIF pathways and subsequently inhibits TLR signaling in innate immune responses.
Collapse
|
36
|
Bánki Z, Posch W, Ejaz A, Oberhauser V, Willey S, Gassner C, Stoiber H, Dittmer U, Dierich MP, Hasenkrug KJ, Wilflingseder D. Complement as an endogenous adjuvant for dendritic cell-mediated induction of retrovirus-specific CTLs. PLoS Pathog 2010; 6:e1000891. [PMID: 20442876 PMCID: PMC2861708 DOI: 10.1371/journal.ppat.1000891] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 04/01/2010] [Indexed: 12/24/2022] Open
Abstract
Previous studies have demonstrated the involvement of complement (C) in induction of efficient CTL responses against different viral infections, but the exact role of complement in this process has not been determined. We now show that C opsonization of retroviral particles enhances the ability of dendritic cells (DCs) to induce CTL responses both in vitro and in vivo. DCs exposed to C-opsonized HIV in vitro were able to stimulate CTLs to elicit antiviral activity significantly better than non-opsonized HIV. Furthermore, experiments using the Friend virus (FV) mouse model illustrated that the enhancing role of complement on DC-mediated CTL induction also occurred in vivo. Our results indicate that complement serves as natural adjuvant for DC-induced expansion and differentiation of specific CTLs against retroviruses.
Collapse
Affiliation(s)
- Zoltán Bánki
- Department of Hygiene, Microbiology and Social Medicine, Division of Virology, Innsbruck Medical University, Innsbruck, Tirol, Austria
| | - Wilfried Posch
- Department of Hygiene, Microbiology and Social Medicine, Division of Virology, Innsbruck Medical University, Innsbruck, Tirol, Austria
| | - Asim Ejaz
- Department of Hygiene, Microbiology and Social Medicine, Division of Virology, Innsbruck Medical University, Innsbruck, Tirol, Austria
| | - Verena Oberhauser
- Department of Hygiene, Microbiology and Social Medicine, Division of Virology, Innsbruck Medical University, Innsbruck, Tirol, Austria
| | - Suzanne Willey
- MRC/UCL Centre for Medical Molecular Virology, Division of Infection and Immunity, University College London, London, United Kingdom
| | - Christoph Gassner
- Central Institute for Blood Transfusion & Immunological Department, Innsbruck, Tirol, Austria
| | - Heribert Stoiber
- Department of Hygiene, Microbiology and Social Medicine, Division of Virology, Innsbruck Medical University, Innsbruck, Tirol, Austria
| | - Ulf Dittmer
- Institute of Virology, University of Duisburg-Essen, Essen, Germany
| | - Manfred P. Dierich
- Department of Hygiene, Microbiology and Social Medicine, Division of Virology, Innsbruck Medical University, Innsbruck, Tirol, Austria
| | - Kim J. Hasenkrug
- Laboratory of Persistent Viral Diseases, Rocky Mountain Laboratories, Hamilton, Montana, United States of America
| | - Doris Wilflingseder
- Department of Hygiene, Microbiology and Social Medicine, Division of Virology, Innsbruck Medical University, Innsbruck, Tirol, Austria
- * E-mail:
| |
Collapse
|
37
|
Wismar R, Brix S, Frøkiaer H, Laerke HN. Dietary fibers as immunoregulatory compounds in health and disease. Ann N Y Acad Sci 2010; 1190:70-85. [PMID: 20388138 DOI: 10.1111/j.1749-6632.2009.05256.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Many nonstarch polysaccharides (NSPs) classified as dietary fibers have been reported to possess immunoregulatory properties. The fibers reported to activate or by other means modulate immune responses originate from both plant, fungal, and microbial sources and constitute highly distinct structures. In order to enhance our understanding of factors important for the immunoregulatory activities, this article addresses the importance of chemical structure, origin, and purity of fibers for their capacity to interact with key regulatory immune cells. Furthermore, we assess bioavailability, and discuss possible mechanisms involved. The binding of some NSPs to carbohydrate receptors on immune cells is well established and this event leads to activation or other changes. Especially, certain beta-glucans and some mannans have demonstrated immunomodulatory capacity with the specific structure being important for the activity. Within beta-glucans the activity varies according to structure, molecular weight, and solubility. As many of the preparations tested constitute crude extracts or partly purified NSPs, the risk of contaminants holding immunoregulatory activities should not be ignored. To what extent NSPs enter systemic circulation has been difficult to assess, partly due to lack of sensitive analytical methods. The presence of NSPs in blood and Peyer's patches in the gut has been demonstrated, supporting encounter between NSPs and immune cells, but bioavailability studies still constitute a major challenge. Studies demonstrating in vivo effects of beta-glucans on microbial infections and cancer treatment strongly indicate an immunoregulatory mechanism behind the effects. However, the potential of NSPs as immunoregulatory food ingredients is still far from fully explored.
Collapse
Affiliation(s)
- René Wismar
- Nutritional Immunology Group, Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | | | | | | |
Collapse
|
38
|
Wang L, Gordon RA, Huynh L, Su X, Park Min KH, Han J, Arthur JS, Kalliolias GD, Ivashkiv LB. Indirect inhibition of Toll-like receptor and type I interferon responses by ITAM-coupled receptors and integrins. Immunity 2010; 32:518-30. [PMID: 20362473 DOI: 10.1016/j.immuni.2010.03.014] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2009] [Revised: 11/25/2009] [Accepted: 03/19/2010] [Indexed: 01/27/2023]
Abstract
An important function of immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors is cross-regulation of heterologous receptor signaling, but mechanisms of cross-inhibition are poorly understood. We show that high-avidity ligation of ITAM-coupled beta2 integrins and FcgammaRs in macrophages inhibited type I interferon receptor and Toll-like receptor (TLR) signaling and induced expression of interleukin-10 (IL-10); signaling inhibitors SOCS3, ABIN-3, and A20; and repressors of cytokine gene transcription STAT3 and Hes1. Induction of inhibitors was dependent on a pathway composed of signaling molecules DAP12, Syk, and Pyk2 that coupled to downstream kinases p38 and MSKs and required integration of IL-10-dependent and -independent signals. ITAM-induced inhibitors abrogated TLR responses by cooperatively targeting distinct steps in TLR signaling. Inhibitory signaling was suppressed by IFN-gamma and attenuated in inflammatory arthritis synovial macrophages. These results provide an indirect mechanism of cross-inhibition of TLRs and delineate a signaling pathway important for deactivation of macrophages.
Collapse
Affiliation(s)
- Lu Wang
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Galicia G, Maes W, Verbinnen B, Kasran A, Bullens D, Arredouani M, Ceuppens JL. Haptoglobin deficiency facilitates the development of autoimmune inflammation. Eur J Immunol 2010; 39:3404-12. [PMID: 19795414 DOI: 10.1002/eji.200939291] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Haptoglobin (HP) is an acute phase protein synthesized by liver cells in response to IL-6. HP has been demonstrated to modulate the immune response and to have anti-inflammatory activities. To analyze HP's effect on autoimmune inflammation, we here studied the course of EAE induced by immunization of Hp knockout (Hp(-/-)) and syngeneic WT mice with myelin oligodendrocyte glycoprotein peptide (MOG(35-55)). Hp(-/-)mice suffered from a more severe disease that was associated with increased expression of IL-17A, IL-6, and IFN-gamma mRNA in the CNS and with a denser cellular infiltrate in the spinal cord. During the recovery phase, a significantly higher number of myeloid DC, CD8+ cells, IL-17+ CD4+ and IFN-gamma+ CD4+ cells persisted in the CNS of Hp(-/-) mice. Absence of HP affected the priming and differentiation of T cells after MOG(35-55) immunization, as levels of Th2 cytokines produced in response to MOG stimulation by Hp(-/-) T cells were reduced. These results suggest that HP plays a modulatory and protective role on autoimmune inflammation of the CNS.
Collapse
Affiliation(s)
- Georgina Galicia
- Division of Clinical Immunology, Gasthuisberg University Hospital, Catholic University of Leuven, Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
40
|
Podgrabinska S, Kamalu O, Mayer L, Shimaoka M, Snoeck H, Randolph GJ, Skobe M. Inflamed lymphatic endothelium suppresses dendritic cell maturation and function via Mac-1/ICAM-1-dependent mechanism. THE JOURNAL OF IMMUNOLOGY 2009; 183:1767-79. [PMID: 19587009 DOI: 10.4049/jimmunol.0802167] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The lymphatic system is essential for the generation of immune responses by facilitating immune cell trafficking to lymph nodes. Dendritic cells (DCs), the most potent APCs, exit tissues via lymphatic vessels, but the mechanisms of interaction between DCs and the lymphatic endothelium and the potential implications of these interactions for immune responses are poorly understood. In this study, we demonstrate that lymphatic endothelial cells (LECs) modulate the maturation and function of DCs. Direct contact of human monocyte-derived DCs with an inflamed, TNF-alpha-stimulated lymphatic endothelium reduced expression of the costimulatory molecule CD86 by DCs and suppressed the ability of DCs to induce T cell proliferation. These effects were dependent on adhesive interactions between DCs and LECs that were mediated by the binding of Mac-1 on DCs to ICAM-1 on LECs. Importantly, the suppressive effects of the lymphatic endothelium on DCs were observed only in the absence of pathogen-derived signals. In vivo, DCs that migrated to the draining lymph nodes upon inflammatory stimuli, but in the absence of a pathogen, showed increased levels of CD86 expression in ICAM-1-deficient mice. Together, these data demonstrate a direct role of LECs in the modulation of immune response and suggest a function of the lymphatic endothelium in preventing undesired immune reactions in inflammatory conditions.
Collapse
Affiliation(s)
- Simona Podgrabinska
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Chen Y, Park YB, Patel E, Silverman GJ. IgM antibodies to apoptosis-associated determinants recruit C1q and enhance dendritic cell phagocytosis of apoptotic cells. THE JOURNAL OF IMMUNOLOGY 2009; 182:6031-43. [PMID: 19414754 DOI: 10.4049/jimmunol.0804191] [Citation(s) in RCA: 170] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Natural Abs, which arise without known immune exposure, have been described that specifically recognize cells dying from apoptosis, but their role in innate immunity remains poorly understood. Herein, we show that the immune response to neoantigenic determinants on apoptotic thymocytes is dominated by Abs to oxidation-associated Ags, phosphorylcholine (PC), a head group that becomes exposed during programmed cell death, and malondialdehyde (MDA), a reactive aldehyde degradation product of polyunsaturated lipids produced following exposure to reactive oxidation species. While natural Abs to apoptotic cells in naive adult mice were dominated by PC and MDA specificities, the amounts of these Abs were substantially boosted by treatment of mice with apoptotic cells. Moreover, the relative amounts of PC and MDA Abs was affected by V(H) gene inheritance. Ab interactions with apoptotic cells also mediated the recruitment of C1q, which enhanced apoptotic cell phagocytosis by immature dendritic cells. Significantly, IgM Abs to both PC and MDA were primary factors in determining the efficiency of serum-dependent apoptotic cell phagocytosis. Hence, we demonstrate a mechanism by which certain natural Abs that recognize neoantigens on apoptotic cells, in naive mice and those induced by immune exposure to apoptotic cells, can enhance the functional capabilities of immature dendritic cells for phagocytic engulfment of apoptotic cells.
Collapse
Affiliation(s)
- Yifang Chen
- Department of Medicine, Laboratory of B-cell Immunobiology, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | | | |
Collapse
|
42
|
Cathepsin X prevents an effective immune response against Helicobacter pylori infection. Eur J Cell Biol 2009; 88:461-71. [PMID: 19446361 DOI: 10.1016/j.ejcb.2009.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2009] [Revised: 03/16/2009] [Accepted: 03/31/2009] [Indexed: 01/24/2023] Open
Abstract
Cathepsin X, a cysteine protease, has been shown to regulate an immune response by activating beta-2 integrin receptors. In this study we demonstrate its role in regulating the immune response to infection with H. pylori. The level of cathepsin X was determined in THP-1 monocyte cells primed with H. pylori antigens isolated from subjects suffering from gastritis, who had either eradicated or not the disease after the antibiotic therapy. We show that the specific clinical outcome of H. pylori eradication therapy correlates strongly with the membrane expression of cathepsin X in stimulated THP-1 cells, being significantly higher after stimulation with H. pylori strains from those subjects who did not respond to antibiotic therapy. The same antigens elicit a more vigorous immune response, increased expression of MHC II, however trigger inadequate cytokine profile (IFN-gamma and IL-4) to eradicate the pathogen. We propose that cathepsin X mediated activation of beta-2 integrin receptor Mac-1 suppresses the stimulatory signal in the form of cytokines. Cathepsin X co-localizes on the membrane of THP-1 cells with Mac-1 integrin receptor and its inhibition increases homotypic aggregation and mononuclear cell proliferation, events that are associated with low Mac-1 activity. Our study highlights the diversity of the innate immune response to H. pylori antigens leading to either successful eradication of the infection or maintenance of chronic inflammation, revealing cathepsin X location and activity as a regulator of the effectiveness of H. pylori eradication.
Collapse
|
43
|
Zimmerli C, Lee BPL, Palmer G, Gabay C, Adams R, Aurrand-Lions M, Imhof BA. Adaptive immune response in JAM-C-deficient mice: normal initiation but reduced IgG memory. THE JOURNAL OF IMMUNOLOGY 2009; 182:4728-36. [PMID: 19342649 DOI: 10.4049/jimmunol.0803892] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have recently shown that junctional adhesion molecule (JAM)-C-deficient mice have leukocytic pulmonary infiltrates, disturbed neutrophil homeostasis, and increased postnatal mortality. This phenotype was partially rescued when mice were housed in ventilated isolators, suggesting an inability to cope with opportunistic infections. In the present study, we further examined the adaptive immune responses in JAM-C(-/-) mice. We found that murine conventional dendritic cells express in addition to Mac-1 and CD11c also JAM-B as ligand for JAM-C. By in vitro adhesion assay, we show that murine DCs can interact with recombinant JAM-C via Mac-1. However, this interaction does not seem to be necessary for dendritic cell migration and function in vivo, even though JAM-C is highly expressed by lymphatic sinuses of lymph nodes. Nevertheless, upon immunization and boosting with a protein Ag, JAM-C-deficient mice showed decreased persistence of specific circulating Abs although the initial response was normal. Such a phenotype has also been observed in a model of Ag-induced arthritis, showing that specific IgG2a Ab titers are reduced in the serum of JAM-C(-/-) compared with wild-type mice. Taken together, these data suggest that JAM-C deficiency affects the adaptive humoral immune response against pathogens, in addition to the innate immune system.
Collapse
Affiliation(s)
- Claudia Zimmerli
- Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | | | | | | | | | | | | |
Collapse
|
44
|
Villiers C, Chevallet M, Diemer H, Couderc R, Freitas H, Van Dorsselaer A, Marche PN, Rabilloud T. From secretome analysis to immunology: chitosan induces major alterations in the activation of dendritic cells via a TLR4-dependent mechanism. Mol Cell Proteomics 2009; 8:1252-64. [PMID: 19279042 DOI: 10.1074/mcp.m800589-mcp200] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dendritic cells are known to be activated by a wide range of microbial products, leading to cytokine production and increased levels of membrane markers such as major histocompatibility complex class II molecules. Such activated dendritic cells possess the capacity to activate naïve T cells. In the present study we demonstrated that immature dendritic cells secrete both the YM1 lectin and lipocalin-2. By testing the ligands of these two proteins, chitosan and siderophores, respectively, we also demonstrated that chitosan, a degradation product of various fungal and protozoal cell walls, induces an activation of dendritic cells at the membrane level, as shown by the up-regulation of membrane proteins such as class II molecules, CD80 and CD86 via a TLR4-dependent mechanism, but is not able to induce cytokine production. This led to the production of activated dendritic cells unable to stimulate T cells. However, costimulation with other microbial products overcame this partial activation and restored the capacity of these activated dendritic cells to stimulate T cells. In addition, successive stimulation with chitosan and then by lipopolysaccharide induced a dose-dependent change in the cytokinic IL-12/IL-10 balance produced by the dendritic cells.
Collapse
Affiliation(s)
- Christian Villiers
- double daggerINSERM U823 Analytical Immunology of Chronic Pathologies, Institut Albert Bonniot, BP170, 38042 Grenoble, France and Université Joseph Fourier, F-38041 Grenoble, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Rossi Paccani S, Benagiano M, Capitani N, Zornetta I, Ladant D, Montecucco C, D'Elios MM, Baldari CT. The adenylate cyclase toxins of Bacillus anthracis and Bordetella pertussis promote Th2 cell development by shaping T cell antigen receptor signaling. PLoS Pathog 2009; 5:e1000325. [PMID: 19266022 PMCID: PMC2643477 DOI: 10.1371/journal.ppat.1000325] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Accepted: 02/03/2009] [Indexed: 01/28/2023] Open
Abstract
The adjuvanticity of bacterial adenylate cyclase toxins has been ascribed to their capacity, largely mediated by cAMP, to modulate APC activation, resulting in the expression of Th2–driving cytokines. On the other hand, cAMP has been demonstrated to induce a Th2 bias when present during T cell priming, suggesting that bacterial cAMP elevating toxins may directly affect the Th1/Th2 balance. Here we have investigated the effects on human CD4+ T cell differentiation of two adenylate cyclase toxins, Bacillus anthracis edema toxin (ET) and Bordetella pertussis CyaA, which differ in structure, mode of cell entry, and subcellular localization. We show that low concentrations of ET and CyaA, but not of their genetically detoxified adenylate cyclase defective counterparts, potently promote Th2 cell differentiation by inducing expression of the master Th2 transcription factors, c-maf and GATA-3. We also present evidence that the Th2–polarizing concentrations of ET and CyaA selectively inhibit TCR–dependent activation of Akt1, which is required for Th1 cell differentiation, while enhancing the activation of two TCR–signaling mediators, Vav1 and p38, implicated in Th2 cell differentiation. This is at variance from the immunosuppressive toxin concentrations, which interfere with the earliest step in TCR signaling, activation of the tyrosine kinase Lck, resulting in impaired CD3ζ phosphorylation and inhibition of TCR coupling to ZAP-70 and Erk activation. These results demonstrate that, notwithstanding their differences in their intracellular localization, which result in focalized cAMP production, both toxins directly affect the Th1/Th2 balance by interfering with the same steps in TCR signaling, and suggest that their adjuvanticity is likely to result from their combined effects on APC and CD4+ T cells. Furthermore, our results strongly support the key role of cAMP in the adjuvanticity of these toxins. Colonization by pathogens requires keeping at bay the host immune defenses, at least at the onset of infection. The adenylate cyclase (AC) toxins produced by many pathogenic bacteria assist in this crucial function by catalyzing the production of cAMP, which acts as a potent immunosuppressant. Nevertheless, at low concentrations, these toxins act as adjuvants, enhancing antibody responses to vaccination. We have investigated the molecular basis of the immunomodulatory activities of two AC toxins, Bacillus anthracis edema toxin and Bordetella pertussis CyaA. We show that high toxin concentrations inhibit activation of T lymphocytes, which orchestrate the adaptive immune response against pathogens, whereas low toxin concentrations promote differentiation of helper T lymphocytes to Th2 effectors, which are required for development of antibody-producing cells. Both the immunosuppressant and Th2–driving activities of the toxins are dependent on cAMP. The results demonstrate that, dependent on their concentration, the AC toxins of B. anthracis and B. pertussis evoke distinct responses on target T lymphocytes by differentially modulating antigen receptor signaling, resulting either in suppression of T cell activation or Th2 cell differentiation. These results are of relevance to the evolution of disease in infected individuals and provide novel mechanistic insight into the adjuvanticity of these toxins.
Collapse
Affiliation(s)
| | - Marisa Benagiano
- Department of Internal Medicine and Immunoallergology, University of Florence, Florence, Italy
| | - Nagaja Capitani
- Department of Evolutionary Biology, University of Siena, Siena, Italy
| | - Irene Zornetta
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Daniel Ladant
- Unité de Biochimie des Interactions Macromoléculaires, CNRS URA 2185, Institut Pasteur, Paris, France
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Mario M. D'Elios
- Department of Internal Medicine and Immunoallergology, University of Florence, Florence, Italy
| | - Cosima T. Baldari
- Department of Evolutionary Biology, University of Siena, Siena, Italy
- * E-mail:
| |
Collapse
|
46
|
Behrens EM, Ning Y, Muvarak N, Zoltick PW, Flake AW, Gallucci S. Apoptotic cell-mediated immunoregulation of dendritic cells does not require iC3b opsonization. THE JOURNAL OF IMMUNOLOGY 2008; 181:3018-26. [PMID: 18713972 DOI: 10.4049/jimmunol.181.5.3018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A number of recent studies show that activation of CR3 on dendritic cells (DCs) suppresses TLR-induced TNF-alpha and IL-12 production and inhibits effective Ag presentation. Although the proposed physiologic role for these phenomena is immune suppression due to recognition of iC3b opsonized apoptotic cells by CR3, all of the aforementioned investigations used artificial means of activating CR3. We investigated whether iC3b opsonized apoptotic cells could induce the same changes reported with artificial ligands such as mAbs or iC3b-opsonized RBC. We explored the kinetics of iC3b opsonization in two models of murine cell apoptosis, gamma-irradiated thymocytes and cytokine deprivation of the IL-3 dependent cell line BaF3. Using a relatively homogenous population of early apoptotic cells (IL-3 deprived BaF3 cells), we show that iC3b opsonized apoptotic cells engage CR3, but this interaction is dispensable in mediating the anti-inflammatory effects of apoptotic cells. TLR-induced TNF-alpha and IL-12 production by bone marrow-derived DCs occurs heterogeneously, with apoptotic cells inhibiting only certain populations depending on the TLR agonist. In contrast, although apoptotic cells induced homogeneous IL-10 production by DCs, IL-10 was not necessary for the inhibition of TNF-alpha and IL-12. Furthermore, because the ability of iC3b opsonization to enhance phagocytosis of apoptotic cells has been controversial, we report that iC3b opsonization does not significantly affect apoptotic cell ingestion by DCs. We conclude that the apoptotic cell receptor system on DCs is sufficiently redundant such that the absence of CR3 engagement does not significantly affect the normal anti-inflammatory processing of apoptotic cells.
Collapse
Affiliation(s)
- Edward M Behrens
- Laboratory of Dendritic Cell Biology, Division of Rheumatology, Joseph Stokes, Jr. Research Institute, Children's Hospital of Philadelphia, Philadelphia, PA 19104-4318, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Spirig R, van Kooten C, Obregon C, Nicod L, Daha M, Rieben R. The complement inhibitor low molecular weight dextran sulfate prevents TLR4-induced phenotypic and functional maturation of human dendritic cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:878-90. [PMID: 18606639 DOI: 10.4049/jimmunol.181.2.878] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Low molecular weight dextran sulfate (DXS) has been reported to inhibit the classical, alternative pathway as well as the mannan-binding lectin pathway of the complement system. Furthermore, it acts as an endothelial cell protectant inhibiting complement-mediated endothelial cell damage. Endothelial cells are covered with a layer of heparan sulfate (HS), which is rapidly released under conditions of inflammation and tissue injury. Soluble HS induces maturation of dendritic cells (DC) via TLR4. In this study, we show the inhibitory effect of DXS on human DC maturation. DXS significantly prevents phenotypic maturation of monocyte-derived DC and peripheral myeloid DC by inhibiting the up-regulation of CD40, CD80, CD83, CD86, ICAM-1, and HLA-DR and down-regulates DC-SIGN in response to HS or exogenous TLR ligands. DXS also inhibits the functional maturation of DC as demonstrated by reduced T cell proliferation, and strongly impairs secretion of the proinflammatory mediators IL-1beta, IL-6, IL-12p70, and TNF-alpha. Exposure to DXS leads to a reduced production of the complement component C1q and a decreased phagocytic activity, whereas C3 secretion is increased. Moreover, DXS was found to inhibit phosphorylation of IkappaB-alpha and activation of NF-kappaB. These findings suggest that DXS prevents TLR-induced maturation of human DC and may therefore be a useful reagent to impede the link between innate and adaptive immunity.
Collapse
Affiliation(s)
- Rolf Spirig
- Departmentof Clinical Research, Laboratory of Cardiovascular Surgery, University of Bern, Bern, Switzerland
| | | | | | | | | | | |
Collapse
|
48
|
Mitchell DA, Ilyas R, Dodds AW, Sim RB. Enzyme-independent, orientation-selective conjugation of whole human complement C3 to protein surfaces. J Immunol Methods 2008; 337:49-54. [DOI: 10.1016/j.jim.2008.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2008] [Accepted: 05/14/2008] [Indexed: 11/26/2022]
|
49
|
Tang ML, Vararattanavech A, Tan SM. Urokinase-type plasminogen activator receptor induces conformational changes in the integrin alphaMbeta2 headpiece and reorientation of its transmembrane domains. J Biol Chem 2008; 283:25392-25403. [PMID: 18644795 DOI: 10.1074/jbc.m802311200] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The glycosylphosphatidylinositol-linked urokinase-type plasminogen activator receptor (uPAR) interacts with the heterodimer cell adhesion molecules integrins to modulate cell adhesion and migration. Devoid of a cytoplasmic domain, uPAR triggers intracellular signaling via its associated molecules that contain cytoplasmic domains. Interestingly, uPAR changes the ectodomain conformation of one of its partner molecules, integrin alpha(5)beta(1), and elicits cytoplasmic signaling. The separation or reorientation of integrin transmembrane domains and cytoplasmic tails are required for integrin outside-in signaling. However, there is a lack of direct evidence showing these conformational changes of an integrin that interacts with uPAR. In this investigation we used reporter monoclonal antibodies and fluorescence resonance energy transfer analyses to show conformational changes in the alpha(M)beta(2) headpiece and reorientation of its transmembrane domains when alpha(M)beta(2) interacts with uPAR.
Collapse
Affiliation(s)
- Man-Li Tang
- Division of Molecular and Cell Biology, School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Ardcharaporn Vararattanavech
- Division of Molecular and Cell Biology, School of Biological Sciences, Nanyang Technological University, 637551, Singapore
| | - Suet-Mien Tan
- Division of Molecular and Cell Biology, School of Biological Sciences, Nanyang Technological University, 637551, Singapore.
| |
Collapse
|
50
|
Cummings KL, Waggoner SN, Tacke R, Hahn YS. Role of complement in immune regulation and its exploitation by virus. Viral Immunol 2008; 20:505-24. [PMID: 18158725 DOI: 10.1089/vim.2007.0061] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Complement is activated during the early phase of viral infection and promotes destruction of virus particles as well as the initiation of inflammatory responses. Recently, complement and complement receptors have been reported to play an important role in the regulation of innate as well as adaptive immune responses during infection. The regulation of host immune responses by complement involves modulation of dendritic cell activity in addition to direct effects on T-cell function. Intriguingly, many viruses encode homologs of complement regulatory molecules or proteins that interact with complement receptors on antigen-presenting cells and lymphocytes. The evolution of viral mechanisms to alter complement function may augment pathogen persistence and limit immune-mediated tissue destruction. These observations suggest that complement may play an important role in both innate and adaptive immune responses to infection as well as virus-mediated modulation of host immunity.
Collapse
Affiliation(s)
- Kara L Cummings
- Beirne Carter Center for Immunology Research and Department of Microbiology, University of Virginia, Charlottesville, Virginia
| | | | | | | |
Collapse
|