1
|
Hölken JM, Teusch N. The Monocytic Cell Line THP-1 as a Validated and Robust Surrogate Model for Human Dendritic Cells. Int J Mol Sci 2023; 24:1452. [PMID: 36674966 PMCID: PMC9866978 DOI: 10.3390/ijms24021452] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
We have implemented an improved, cost-effective, and highly reproducible protocol for a simple and rapid differentiation of the human leukemia monocytic cell line THP-1 into surrogates for immature dendritic cells (iDCs) or mature dendritic cells (mDCs). The successful differentiation of THP-1 cells into iDCs was determined by high numbers of cells expressing the DC activation markers CD54 (88%) and CD86 (61%), and the absence of the maturation marker CD83. The THP-1-derived mDCs are characterized by high numbers of cells expressing CD54 (99%), CD86 (73%), and the phagocytosis marker CD11b (49%) and, in contrast to THP-1-derived iDCs, CD83 (35%) and the migration marker CXCR4 (70%). Treatment of iDCs with sensitizers, such as NiSO4 and DNCB, led to high expression of CD54 (97%/98%; GMFI, 3.0/3.2-fold induction) and CD86 (64%/96%; GMFI, 4.3/3.2-fold induction) compared to undifferentiated sensitizer-treated THP-1 (CD54, 98%/98%; CD86, 55%/96%). Thus, our iDCs are highly suitable for toxicological studies identifying potential sensitizing or inflammatory compounds. Furthermore, the expression of CD11b, CD83, and CXCR4 on our iDC and mDC surrogates could allow studies investigating the molecular mechanisms of dendritic cell maturation, phagocytosis, migration, and their use as therapeutic targets in various disorders, such as sensitization, inflammation, and cancer.
Collapse
Affiliation(s)
| | - Nicole Teusch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| |
Collapse
|
2
|
Waad Sadiq Z, Brioli A, Al-Abdulla R, Çetin G, Schütt J, Murua Escobar H, Krüger E, Ebstein F. Immunogenic cell death triggered by impaired deubiquitination in multiple myeloma relies on dysregulated type I interferon signaling. Front Immunol 2023; 14:982720. [PMID: 36936919 PMCID: PMC10018035 DOI: 10.3389/fimmu.2023.982720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 02/06/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Proteasome inhibition is first line therapy in multiple myeloma (MM). The immunological potential of cell death triggered by defects of the ubiquitin-proteasome system (UPS) and subsequent perturbations of protein homeostasis is, however, less well defined. Methods In this paper, we applied the protein homeostasis disruptors bortezomib (BTZ), ONX0914, RA190 and PR619 to various MM cell lines and primary patient samples to investigate their ability to induce immunogenic cell death (ICD). Results Our data show that while BTZ treatment triggers sterile type I interferon (IFN) responses, exposure of the cells to ONX0914 or RA190 was mostly immunologically silent. Interestingly, inhibition of protein de-ubiquitination by PR619 was associated with the acquisition of a strong type I IFN gene signature which relied on key components of the unfolded protein and integrated stress responses including inositol-requiring enzyme 1 (IRE1), protein kinase R (PKR) and general control nonderepressible 2 (GCN2). The immunological relevance of blocking de-ubiquitination in MM was further reflected by the ability of PR619-induced apoptotic cells to facilitate dendritic cell (DC) maturation via type I IFN-dependent mechanisms. Conclusion Altogether, our findings identify de-ubiquitination inhibition as a promising strategy for inducing ICD of MM to expand current available treatments.
Collapse
Affiliation(s)
- Zeinab Waad Sadiq
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| | - Annamaria Brioli
- Klinik und Poliklinik für Innere Medizin C, Universitätsmedizin Greifswald, Greifswald, Germany
- Klinik für Innere Medizin II, Universitätsklinikum Jena, Jena, Germany
| | - Ruba Al-Abdulla
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| | - Gonca Çetin
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| | - Jacqueline Schütt
- Klinik und Poliklinik für Innere Medizin C, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Hugo Murua Escobar
- Department of Medicine, Clinic III, Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, Rostock, Germany
| | - Elke Krüger
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| | - Frédéric Ebstein
- Institut für Medizinische Biochemie und Molekularbiologie (IMBM), Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
3
|
Li S, Wang D, Cheng J, Sun J, Kalvakolanu DV, Zhao X, Wang D, You Y, Zhang L, Yu D. A photodynamically sensitized dendritic cell vaccine that promotes the anti-tumor effects of anti-PD-L1 monoclonal antibody in a murine model of head and neck squamous cell carcinoma. J Transl Med 2022; 20:505. [PMID: 36329529 PMCID: PMC9635135 DOI: 10.1186/s12967-022-03707-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors are promising tools in combating several cancers, including head and neck squamous cell carcinoma (HNSCC). However, a substantial portion of HNSCC patients do not respond to PD-L1 antibody. Here we describe a photodynamic therapeutic (PDT) approach to enhance anti-tumor effects of the anti-PD-L1 antibody. METHODS Phototoxicity of PDT was confirmed using fluorescence microscopy, Cell Counting Kit-8 (CCK-8), Enzyme Linked Immunosorbent Assay (ELISA) and flow cytometry analyses. Phenotypic and functional maturation of immature DCs (imDCs) induced by PDT were measured using flow cytometry and ELISA. A mouse model was established using the HNSCC line, SCC7, and was used to evaluate therapeutic effects of PDT-DC vaccine in facilitating anti-tumor immunity of PD-L1 antibody. RESULTS Immunogenic cell death (ICD) of SCC7 cells was induced by PDT with 0.5 µM of m-THPC and the 5 J/cm2 of light dose. ICD of SCC7 cells stimulated imDCs maturation. In vivo assays suggested that PDT-DC vaccine and anti-PD-L1 mAb synergistically induced anti-tumor immunity and suppressed tumor progression. CONCLUSION PDT-DC vaccine enhances therapeutic effects of PD-L1 antibody, which might provide a novel approach for HNSCC immunotherapy.
Collapse
Affiliation(s)
- Shuang Li
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, 130041, Changchun, Jilin Province, People's Republic of China
| | - Ding Wang
- Key Laboratory of Pathobiology, Department of pathophysiology, College of Basic Medical Sciences, Ministry of Education, Jilin University, 126 Xinmin Street, 130012, Changchun, Jilin, P.R. China
| | - Jinzhang Cheng
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, 130041, Changchun, Jilin Province, People's Republic of China
| | - Jicheng Sun
- Key Laboratory of Pathobiology, Department of pathophysiology, College of Basic Medical Sciences, Ministry of Education, Jilin University, 126 Xinmin Street, 130012, Changchun, Jilin, P.R. China
| | - Dhan V Kalvakolanu
- Key Laboratory of Pathobiology, Department of pathophysiology, College of Basic Medical Sciences, Ministry of Education, Jilin University, 126 Xinmin Street, 130012, Changchun, Jilin, P.R. China.,Greenebaum NCI Comprehensive Cancer Center, Department of Microbiology and Immunology, University of Maryland School Medicine, Baltimore, MD, USA
| | - Xue Zhao
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, 130041, Changchun, Jilin Province, People's Republic of China
| | - Di Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, 130041, Changchun, Jilin Province, People's Republic of China
| | - Yunhan You
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, 130041, Changchun, Jilin Province, People's Republic of China
| | - Ling Zhang
- Key Laboratory of Pathobiology, Department of pathophysiology, College of Basic Medical Sciences, Ministry of Education, Jilin University, 126 Xinmin Street, 130012, Changchun, Jilin, P.R. China.
| | - Dan Yu
- Department of Otolaryngology-Head and Neck Surgery, The Second Hospital of Jilin University, No. 218, Ziqiang Street, Nanguan District, 130041, Changchun, Jilin Province, People's Republic of China.
| |
Collapse
|
4
|
Hong S, Park YH, Lee J, Moon J, Kong E, Jeon J, Park JC, Kim HR, Kim P. 3D Visualization of Dynamic Cellular Reaction of Pulpal CD11c+ Dendritic Cells against Pulpitis in Whole Murine Tooth. Int J Mol Sci 2021; 22:ijms222312683. [PMID: 34884488 PMCID: PMC8657593 DOI: 10.3390/ijms222312683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 11/16/2022] Open
Abstract
In dental pulp, diverse types of cells mediate the dental pulp immunity in a highly complex and dynamic manner. Yet, 3D spatiotemporal changes of various pulpal immune cells dynamically reacting against foreign pathogens during immune response have not been well characterized. It is partly due to the technical difficulty in detailed 3D comprehensive cellular-level observation of dental pulp in whole intact tooth beyond the conventional histological analysis using thin tooth slices. In this work, we validated the optical clearing technique based on modified Murray’s clear as a valuable tool for a comprehensive cellular-level analysis of dental pulp. Utilizing the optical clearing, we successfully achieved a 3D visualization of CD11c+ dendritic cells in the dentin-pulp complex of a whole intact murine tooth. Notably, a small population of unique CD11c+ dendritic cells extending long cytoplasmic processes into the dentinal tubule while located at the dentin-pulp interface like odontoblasts were clearly visualized. 3D visualization of whole murine tooth enabled a reliable observation of these rarely existing cells with a total number less than a couple of tens in one tooth. These CD11c+ dendritic cells with processes in the dentinal tubule were significantly increased in the dental pulpitis model induced by mechanical and chemical irritation. Additionally, the 3D visualization revealed a distinct spatial 3D arrangement of pulpal CD11c+ cells in the pulp into a front-line barrier-like formation in the pulp within 12 h after the irritation. Collectively, these observations demonstrated the unique capability of optical clearing-based comprehensive 3D cellular-level visualization of the whole tooth as an efficient method to analyze 3D spatiotemporal changes of various pulpal cells in normal and pathological conditions.
Collapse
Affiliation(s)
- Sujung Hong
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea; (S.H.); (J.L.); (J.M.)
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon 34141, Korea; (E.K.); (J.J.)
| | - Yeoung-Hyun Park
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Korea; (Y.-H.P.); (J.-C.P.)
| | - Jingu Lee
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea; (S.H.); (J.L.); (J.M.)
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon 34141, Korea; (E.K.); (J.J.)
| | - Jieun Moon
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea; (S.H.); (J.L.); (J.M.)
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon 34141, Korea; (E.K.); (J.J.)
| | - Eunji Kong
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon 34141, Korea; (E.K.); (J.J.)
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Jehwi Jeon
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon 34141, Korea; (E.K.); (J.J.)
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea
| | - Joo-Cheol Park
- Department of Oral Histology-Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 03080, Korea; (Y.-H.P.); (J.-C.P.)
| | - Hyung-Ryong Kim
- Department of Pharmacology, College of Dentistry, Jeonbuk National University, Jeonju 54896, Korea
- Correspondence: (H.-R.K.); (P.K.)
| | - Pilhan Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea; (S.H.); (J.L.); (J.M.)
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon 34141, Korea; (E.K.); (J.J.)
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Korea
- Correspondence: (H.-R.K.); (P.K.)
| |
Collapse
|
5
|
Abstract
Dendritic cells (DCs) are efficient antigen-presenting cells that serve as a link between the innate and adaptive immune systems. These cells are broadly involved in cellular and humoral immune responses by presenting antigens to initiate T cell reactions, cytokine and chemokine secretion, T cell differentiation and expansion, B cell activation and regulation, and the mediation of immune tolerance. The functions of DCs depend on their activation status, which is defined by the stages of maturation, phenotype differentiation, and migration ability, among other factors. IL-6 is a soluble mediator mainly produced by a variety of immune cells, including DCs, that exerts pleiotropic effects on immune and inflammatory responses through interaction with specific receptors expressed on the surface of target cells. Here, we review the role of IL-6, when generated in an inflammatory context or as derived from DCs, in modulating the biologic function and activation status of DCs and emphasize the importance of searching for novel strategies to target the IL-6/IL-6 signaling pathway as a means to diminish the inflammatory activity of DCs in immune response or to prime the immunogenic activity of DCs in immunosuppressive conditions.
Collapse
Affiliation(s)
- Yu-Dong Xu
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mi Cheng
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pan-Pan Shang
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Qing Yang
- Shanghai Research Institute of Acupuncture and Meridian, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Husain I, Luo X. Apoptotic Donor Cells in Transplantation. Front Immunol 2021; 12:626840. [PMID: 33717145 PMCID: PMC7947657 DOI: 10.3389/fimmu.2021.626840] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/18/2021] [Indexed: 12/31/2022] Open
Abstract
Despite significant advances in prevention and treatment of transplant rejection with immunosuppressive medications, we continue to face challenges of long-term graft survival, detrimental medication side effects to both the recipient and transplanted organ together with risks for opportunistic infections. Transplantation tolerance has so far only been achieved through hematopoietic chimerism, which carries with it a serious and life-threatening risk of graft versus host disease, along with variability in persistence of chimerism and uncertainty of sustained tolerance. More recently, numerous in vitro and in vivo studies have explored the therapeutic potential of silent clearance of apoptotic cells which have been well known to aid in maintaining peripheral tolerance to self. Apoptotic cells from a donor not only have the ability of down regulating the immune response, but also are a way of providing donor antigens to recipient antigen-presenting-cells that can then promote donor-specific peripheral tolerance. Herein, we review both laboratory and clinical evidence that support the utility of apoptotic cell-based therapies in prevention and treatment of graft versus host disease and transplant rejection along with induction of donor-specific tolerance in solid organ transplantation. We have highlighted the potential limitations and challenges of this apoptotic donor cell-based therapy together with ongoing advancements and attempts made to overcome them.
Collapse
Affiliation(s)
- Irma Husain
- Department of Medicine, Duke University, Durham, NC, United States
| | - Xunrong Luo
- Department of Medicine, Duke University, Durham, NC, United States
| |
Collapse
|
7
|
Zhao N, Dong W, Kim H, Moallemian R, Lv J, Wang H, Zheng H, Wei F, Ma X. Capping protein regulator and myosin 1 linker 3 regulates transcription of key cytokines in activated phagocytic cells. Cell Signal 2020; 78:109848. [PMID: 33246003 DOI: 10.1016/j.cellsig.2020.109848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/19/2020] [Accepted: 11/22/2020] [Indexed: 01/02/2023]
Abstract
We have recently reported that capping protein regulator and myosin 1 linker 3 (CARMIL3), first identified as an oncofetal-like gene, is required for metastasis of breast and prostate cancer cells via regulating the actin cytoskeletal dynamics near the plasma membrane. Here, we demonstrate a novel function of CARMIL3 as an essential regulator of the transcription of several key proinflammatory cytokines in macrophages engulfing apoptotic cells and/or exposed to lipopolysaccharides (LPS). CARMIL3-deficient macrophages expressed strongly abrogated levels of interleukin (IL)-6, TNF-α, IL-1β and IL-23 in response to LPS, whereas IL-10 expression was enhanced. An RNA-seq analysis of CARMIL3-deficient and wild-type (WT) RAW264.7 cells stimulated with LPS revealed many differentially expressed genes, impacting several important inflammatory pathways. At the molecular level, CARMIL3 deficiency caused a strong impairment in LPS-activated nuclear factor-κB (NF-κB) signaling with decreased IKKα/β and IκBα phosphorylation and severely reduced p65 protein levels. This study uncovers a crucial role of CARMIL3 in impacting the balance between inflammation and tissue homeostasis via regulating major cytokines production in phagocytic cells.
Collapse
Affiliation(s)
- Na Zhao
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wenjuan Dong
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Hajeong Kim
- Department of Physiology, Kyungpook National University School of Medicine, Daegu 41944, Republic of Korea
| | - Rezvan Moallemian
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jiyang Lv
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Huan Wang
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hua Zheng
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fang Wei
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Xiaojing Ma
- State Key Laboratory of Microbial Metabolism, Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
8
|
Franklin C, Bruderek K, Schilling B, Brandau S. Chemoirradiated neutrophils and T cells differentially affect immune functions of APCs. J Leukoc Biol 2019; 106:481-493. [PMID: 31075186 DOI: 10.1002/jlb.5a0618-242r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 04/02/2019] [Accepted: 04/18/2019] [Indexed: 01/08/2023] Open
Abstract
Extracorporeal photopheresis (ECP) is known as an immunomodulatory therapy with few side effects, which is mainly used in the treatment of cutaneous T cell lymphoma, graft-versus-host disease, and allograft rejection. During ECP, leukocytes are separated from whole blood by leukapheresis, subsequently chemoirradiated with 8-methoxypsoralen and UVA light, and re-infused into the patient. Although clinically effective, its mode of action has not been fully elucidated. In the present study, we analyzed the interaction of chemoirradiated neutrophils and CD3+ lymphocytes with APC in an in vitro model. We report that chemoirradiated CD3+ T cells induced increased expression of activation markers on dendritic cells (DC), macrophages, and monocytes. Coculture of chemoirradiated CD3+ T cells with these APC also led to significantly increased secretion of TNF-α. Although less pronounced, additional activation of APC took place when APC were stimulated with LPS or IFN-γ. In contrast, chemoirradiated neutrophils did not show activating effects on APC. The presence of chemoirradiated neutrophils during LPS and IFN-γ stimulation of DC rather diminished DC and macrophage activation. In line with these findings DC cocultured with chemoirradiated CD3+ T cells, but not neutrophils, showed significantly increased activation of CD3+ responder lymphocytes in a mixed lymphocyte reaction. With this study, we demonstrate that chemoirradiated leukocytes have differential indirect immunomodulatory effects. Whereas chemoirradiated CD3+ T cells activate APC, chemoirradiated neutrophils suppress activation of APC in the presence of other activating factors, suggesting that the composition of the ECP-treated buffy coat might be of importance for its immunomodulatory effects.
Collapse
Affiliation(s)
- Cindy Franklin
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.,Research Division, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany.,Department of Dermatology and Venereology, University Hospital of Cologne, Cologne, Germany
| | - Kirsten Bruderek
- Research Division, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Bastian Schilling
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.,Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Sven Brandau
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany.,German Cancer Consortium (DKTK) Partner Site, Essen-Düsseldorf, Essen, Germany
| |
Collapse
|
9
|
Tumangelova-Yuzeir K, Naydenov E, Ivanova-Todorova E, Krasimirova E, Vasilev G, Nachev S, Kyurkchiev D. Mesenchymal Stem Cells Derived and Cultured from Glioblastoma Multiforme Increase Tregs, Downregulate Th17, and Induce the Tolerogenic Phenotype of Monocyte-Derived Cells. Stem Cells Int 2019; 2019:6904638. [PMID: 31191680 PMCID: PMC6525812 DOI: 10.1155/2019/6904638] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/08/2019] [Accepted: 02/24/2019] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess immunosuppressive properties and have been described in the tumor microenvironment of glioblastoma multiforme (GBM). This manuscript has two major topics-first, to describe isolated and cultured MSCs derived from GBM (GB-MSCs) and second, to examine their in vitro immunosuppressive capacity. Our results display cells with morphology and phenotype, clonogenic ability, and osteogenic potential, typical for MSCs. Furthermore, the cultured cells show intracellular expression of the neural markers Nestin and GFAP. They express PD-L1 and secrete TGFβ, CCL-2, PGE2, IL-6, and sVEGF. Coculturing of GB-MSCs with PBMCs isolated from healthy donors results in a decreased percentage of Th17 lymphocytes and an increased percentage of Tregs. Regarding the impact of GB-MSCs on monocytes, we establish an augmented expression of CD14 and CD86 along with diminished expression of HLA-DR and CD80, which is associated with tolerogenic phenotype monocyte-derived cells. In conclusion, our results describe in detail GBM-derived and cultured cells that meet the criteria for MSCs but at the same time express Nestin and GFAP. GB-MSCs express and secrete suppressive molecules, influencing in vitro T cells and monocytes, and are probably another factor involved in the immune suppression exerted by GBM.
Collapse
Affiliation(s)
- Kalina Tumangelova-Yuzeir
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Emanuil Naydenov
- Clinic of Neurosurgery, University Hospital “St. Ivan Rilski,” Medical University Sofia, 15 “Acad. Ivan Geshov” Str., 1431 Sofia, Bulgaria
| | - Ekaterina Ivanova-Todorova
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Ekaterina Krasimirova
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Georgi Vasilev
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| | - Sevdalin Nachev
- Laboratory of Clinical Pathology, University Hospital “St. Ivan Rilski,” Medical University Sofia, 15 “Acad. Ivan Geshov” Str., 1431 Sofia, Bulgaria
| | - Dobroslav Kyurkchiev
- Laboratory of Clinical Immunology, University Hospital “St. Ivan Rilski,” Department of Clinical Laboratory and Clinical Immunology, Medical University of Sofia, Sofia 1431, Bulgaria
| |
Collapse
|
10
|
Mohammadi Shahrokhi V, Ravari A, Mirzaei T, Zare-Bidaki M, Asadikaram G, Arababadi MK. IL-17A and IL-23: plausible risk factors to induce age-associated inflammation in Alzheimer's disease. Immunol Invest 2018; 47:812-822. [PMID: 30081688 DOI: 10.1080/08820139.2018.1504300] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background: Aging and its complications such as Alzheimer's disease (AD) are associated with chronic low-grade inflammation entitled age-associated inflammation. However, the main mechanisms whichinduce age-associated inflammation in aging and AD are yet to beclarified. L-23/IL-17A axis plays important roles in the induction of inflammation and consequently autoimmune disease. This review evaluates the main roles played by IL-17A, IL-23, and IL-17A/IL-23 axis in the pathogenesis of age-associated inflammation in AD patients. Result: IL-23/IL-17A axis, is an important factor participate in the pathogenesis of age-associated inflammation. The genetic variations and microbial infection can be considered as the most important candidates to induce AD via upregulation of IL-17A. IL-17A also deteriorates AD via induction by amyloid-β. IL-17A participates in the induction of AD by increasing neutrophils infiltration to brain, induction of neuroinflammation, increase in FASL, and amyloid-βdeposition as well as activation of microglia. Conclusions: Due to the important roles played by IL-23/IL-17A axis in AD pathogenesis, it can be considered as a target for immunotherapy against AD. Abbreviations: Aβ: β-Amyloid; AD: Alzheimer's disease; CD: cluster of differentiation; DAMPs: Damage-associated molecular patterns; DCs: dendritic cells; HLA: human leukocyte antigen; NF-κB: nuclear factor kappa-light-chain-enhancer of activated B cells; RAR: retinoic-acid receptor; RORγt: RAR-related orphan receptor gamma t; SAMP8: senescence-accelerated mouse prone 8 strain; TGF-β: tumor growth factor-β; TLRs: toll-like receptors.
Collapse
Affiliation(s)
- Vahid Mohammadi Shahrokhi
- a Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences , Rafsanjan University of Medical Sciences , Rafsanjan , Iran.,b Dept. of Immunology, Faculty of Medicine , Rafsanjan University of Medical Sciences , Rafsanjan , Iran
| | - Ali Ravari
- c Geriatric Care Research Center, Research Institute of Basic Medical Sciences , Rafsanjan , Iran.,d Dept. of Medical Surgical Nursing, Faculty of Nursing and Midwifery , Rafsanjan University of Medical Sciences , Rafsanjan , Iran
| | - Tayebeh Mirzaei
- c Geriatric Care Research Center, Research Institute of Basic Medical Sciences , Rafsanjan , Iran.,d Dept. of Medical Surgical Nursing, Faculty of Nursing and Midwifery , Rafsanjan University of Medical Sciences , Rafsanjan , Iran
| | - Mohammad Zare-Bidaki
- a Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences , Rafsanjan University of Medical Sciences , Rafsanjan , Iran.,e Dept. of Microbiology, Faculty of Medicine , Rafsanjan University of Medical Sciences , Rafsanjan , Iran
| | - Gholamreza Asadikaram
- f Neuroscience Research Center, Institute of Neuropharmacology , Kerman University of Medical Sciences , Kerman , Iran.,g Department of Biochemistry, School of Medicine , Kerman University of Medical Sciences , Kerman , Iran
| | - Mohammad Kazemi Arababadi
- a Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences , Rafsanjan University of Medical Sciences , Rafsanjan , Iran.,b Dept. of Immunology, Faculty of Medicine , Rafsanjan University of Medical Sciences , Rafsanjan , Iran
| |
Collapse
|
11
|
Androulidaki A, Wachsmuth L, Polykratis A, Pasparakis M. Differential role of MyD88 and TRIF signaling in myeloid cells in the pathogenesis of autoimmune diabetes. PLoS One 2018. [PMID: 29522531 PMCID: PMC5844544 DOI: 10.1371/journal.pone.0194048] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Type 1 diabetes (T1D) is caused by the autoimmune destruction of the insulin-producing pancreatic beta cells. While the role of adaptive immunity has been extensively studied, the role of innate immune responses and particularly of Toll- like Receptor (TLR) signaling in T1D remains poorly understood. Here we show that myeloid cell-specific MyD88 deficiency considerably protected mice from the development of streptozotocin (STZ)-induced diabetes. The protective effect of MyD88 deficiency correlated with increased expression of the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO) in pancreatic lymph nodes from STZ-treated mice and in bone marrow-derived dendritic cells (BMDC) stimulated with apoptotic cells. Mice with myeloid cell specific TIR-domain-containing adapter-inducing interferon-β (TRIF) knockout showed a trend towards accelerated onset of STZ-induced diabetes, while TRIF deficiency resulted in reduced IDO expression in vivo and in vitro. Moreover, myeloid cell specific MyD88 deficiency delayed the onset of diabetes in Non-Obese Diabetic (NOD) mice, whereas TRIF deficiency had no effect. Taken together, these results identify MyD88 signaling in myeloid cells as a critical pathogenic factor in autoimmune diabetes, which is antagonized by TRIF-dependent responses. This differential function of MyD88 and TRIF depends at least in part on their opposite effects in regulating IDO expression in phagocytes exposed to apoptotic cells.
Collapse
MESH Headings
- Adaptor Proteins, Vesicular Transport/deficiency
- Adaptor Proteins, Vesicular Transport/genetics
- Adaptor Proteins, Vesicular Transport/physiology
- Animals
- Apoptosis
- Dendritic Cells/physiology
- Diabetes Mellitus, Experimental/etiology
- Diabetes Mellitus, Experimental/immunology
- Diabetes Mellitus, Type 1/etiology
- Diabetes Mellitus, Type 1/immunology
- Enzyme Induction
- Female
- Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Interferon-gamma/biosynthesis
- Interferon-gamma/genetics
- Macrophages, Peritoneal/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Myeloid Cells/immunology
- Myeloid Differentiation Factor 88/deficiency
- Myeloid Differentiation Factor 88/genetics
- Myeloid Differentiation Factor 88/physiology
- Phagocytosis
- Specific Pathogen-Free Organisms
- Streptozocin
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- Ariadne Androulidaki
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Laurens Wachsmuth
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Apostolos Polykratis
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Manolis Pasparakis
- Institute for Genetics, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
- * E-mail:
| |
Collapse
|
12
|
Szondy Z, Sarang Z, Kiss B, Garabuczi É, Köröskényi K. Anti-inflammatory Mechanisms Triggered by Apoptotic Cells during Their Clearance. Front Immunol 2017; 8:909. [PMID: 28824635 PMCID: PMC5539239 DOI: 10.3389/fimmu.2017.00909] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 07/17/2017] [Indexed: 12/19/2022] Open
Abstract
In the human body, billions of cells die by apoptosis every day. The subsequent clearance of apoptotic cells by phagocytosis is normally efficient enough to prevent secondary necrosis and the consequent release of cell contents that would induce inflammation and trigger autoimmunity. In addition, apoptotic cells generally induce an anti-inflammatory response, thus removal of apoptotic cells is usually immunologically silent. Since the first discovery that uptake of apoptotic cells leads to transforming growth factor (TGF)-β and interleukin (IL)-10 release by engulfing macrophages, numerous anti-inflammatory mechanisms triggered by apoptotic cells have been discovered, including release of anti-inflammatory molecules from the apoptotic cells, triggering immediate anti-inflammatory signaling pathways by apoptotic cell surface molecules via phagocyte receptors, activating phagocyte nuclear receptors following uptake and inducing the production of anti-inflammatory soluble mediators by phagocytes that may act via paracrine or autocrine mechanisms to amplify and preserve the anti-inflammatory state. Here, we summarize our present knowledge about how these anti-inflammatory mechanisms operate during the clearance of apoptotic cells.
Collapse
Affiliation(s)
- Zsuzsa Szondy
- Department of Biochemistry and Molecular Biology of Medical Faculty, University of Debrecen, Debrecen, Hungary.,Department of Basic Medical Sciences of Dental Faculty, University of Debrecen, Debrecen, Hungary
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology of Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Beáta Kiss
- Department of Biochemistry and Molecular Biology of Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Éva Garabuczi
- Department of Biochemistry and Molecular Biology of Medical Faculty, University of Debrecen, Debrecen, Hungary
| | - Krisztina Köröskényi
- Department of Biochemistry and Molecular Biology of Medical Faculty, University of Debrecen, Debrecen, Hungary.,Department of Basic Medical Sciences of Dental Faculty, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
13
|
Abstract
Type 1 diabetes (T1D) is a metabolic disease that results from the autoimmune attack against insulin-producing β-cells in the pancreatic islets of Langerhans. Currently, there is no treatment to restore endogenous insulin secretion in patients with autoimmune diabetes. In the last years, the development of new therapies to induce long-term tolerance has been an important medical health challenge. Apoptosis is a physiological mechanism that contributes to the maintenance of immune tolerance. Apoptotic cells are a source of autoantigens that induce tolerance after their removal by antigen presenting cells (APCs) through a process called efferocytosis. Efferocytosis will not cause maturation in dendritic cells, one of the most powerful APCs, and this process could induce tolerance rather than autoimmunity. However, failure of this mechanism due to an increase in the rate of β-cells apoptosis and/or defects in efferocytosis results in activation of APCs, contributing to inflammation and to the loss of tolerance to self. In fact, T1D and other autoimmune diseases are associated to enhanced apoptosis of target cells and defective apoptotic cell clearance. Although further research is needed, the clinical relevance of immunotherapies based on apoptosis could prove to be very important, as it has translational potential in situations that require the reestablishment of immunological tolerance, such as autoimmune diseases. This review summarizes the effects of apoptosis of β-cells towards autoimmunity or tolerance and its application in the field of emerging immunotherapies.
Collapse
|
14
|
Kurosaki T, Nakasone C, Kodama Y, Egashira K, Harasawa H, Muro T, Nakagawa H, Kitahara T, Higuchi N, Nakamura T, Sasaki H. Splenic Gene Delivery System Using Self-assembling Nano-complex with Phosphatidylserine Analog. Biol Pharm Bull 2015; 38:23-9. [DOI: 10.1248/bpb.b14-00478] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
| | | | - Yukinobu Kodama
- Department of Hospital Pharmacy, Nagasaki University Hospital
| | - Kanoko Egashira
- Department of Hospital Pharmacy, Nagasaki University Hospital
| | - Hitomi Harasawa
- Department of Hospital Pharmacy, Nagasaki University Hospital
| | - Takahiro Muro
- Department of Hospital Pharmacy, Nagasaki University Hospital
| | - Hiroo Nakagawa
- Department of Hospital Pharmacy, Nagasaki University Hospital
| | | | | | | | - Hitoshi Sasaki
- Department of Hospital Pharmacy, Nagasaki University Hospital
| |
Collapse
|
15
|
Ruiz P, Maldonado P, Hidalgo Y, Gleisner A, Sauma D, Silva C, Saez JJ, Nuñez S, Rosemblatt M, Bono MR. Transplant tolerance: new insights and strategies for long-term allograft acceptance. Clin Dev Immunol 2013; 2013:210506. [PMID: 23762087 PMCID: PMC3665173 DOI: 10.1155/2013/210506] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 04/12/2013] [Accepted: 04/13/2013] [Indexed: 02/08/2023]
Abstract
One of the greatest advances in medicine during the past century is the introduction of organ transplantation. This therapeutic strategy designed to treat organ failure and organ dysfunction allows to prolong the survival of many patients that are faced with no other treatment option. Today, organ transplantation between genetically dissimilar individuals (allogeneic grafting) is a procedure widely used as a therapeutic alternative in cases of organ failure, hematological disease treatment, and some malignancies. Despite the potential of organ transplantation, the administration of immunosuppressive drugs required for allograft acceptance induces severe immunosuppression in transplanted patients, which leads to serious side effects such as infection with opportunistic pathogens and the occurrence of neoplasias, in addition to the known intrinsic toxicity of these drugs. To solve this setback in allotransplantation, researchers have focused on manipulating the immune response in order to create a state of tolerance rather than unspecific immunosuppression. Here, we describe the different treatments and some of the novel immunotherapeutic strategies undertaken to induce transplantation tolerance.
Collapse
Affiliation(s)
- Paulina Ruiz
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
- Programa de Ciencias Biomedicas, Facultad de Medicina, Universidad de Chile, 8380453 Santiago, Chile
| | - Paula Maldonado
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
| | - Yessia Hidalgo
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
| | - Alejandra Gleisner
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
| | - Daniela Sauma
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
- Fundacion Ciencia y Vida, 7780272 Santiago, Chile
| | - Cinthia Silva
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
| | - Juan Jose Saez
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
| | - Sarah Nuñez
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
| | - Mario Rosemblatt
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
- Fundacion Ciencia y Vida, 7780272 Santiago, Chile
- Facultad de Ciencias Biologicas, Universidad Andres Bello, 8370146 Santiago, Chile
| | - Maria Rosa Bono
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, 7800024 Santiago, Chile
| |
Collapse
|
16
|
SIGN-R1, a C-type lectin, enhances apoptotic cell clearance through the complement deposition pathway by interacting with C1q in the spleen. Cell Death Differ 2012; 20:535-45. [PMID: 23238564 DOI: 10.1038/cdd.2012.160] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Complements, such as C1q and C3, and macrophages in the splenic marginal zone (MZMs) play pivotal roles in the efficient uptake and processing of circulating apoptotic cells. SIGN-R1, a C-type lectin that is highly expressed in a subpopulation of MZMs, regulates the complement fixation pathway by interacting with C1q, to fight blood-borne Streptococcus pneumoniae. Therefore, we examined whether the SIGN-R1-mediated classical complement pathway plays a role in apoptotic cell clearance and immune tolerance. SIGN-R1 first-bound apoptotic cells and this binding was significantly enhanced in the presence of C1q. SIGN-R1-C1q complex then immediately mediated C3 deposition on circulating apoptotic cells in the MZ, leading to the efficient clearance of them. SIGN-R1-mediated C3 deposition was completely abolished in the spleen of SIGN-R1 knockout (KO) mice. Given that SIGN-R1 is not expressed in the liver, we were struck by the finding that C3-deposited apoptotic cells were still found in the liver of wild-type mice, and dramatically reduced in the SIGN-R1 KO liver. In particular, SIGN-R1 deficiency caused delayed clearance of apoptotic cells and aberrant secretion of cytokines, such as TNF-α, IL-6, and TGF-β in the spleen as well as in the liver. In addition, anti-double- and single-stranded DNA antibody level was significantly increased in SIGN-R1-depleted mice compared with control mice. These findings suggest a novel mechanism of apoptotic cell clearance which is initiated by SIGN-R1 in the MZ and identify an integrated role of SIGN-R1 in the systemic clearance of apoptotic cells, linking the recognition of apoptotic cells, the opsonization of complements, and the induction of immune tolerance.
Collapse
|
17
|
Valentin-Torres A, Ramirez Kitchen CM, Haller HS, Bernstein HB. Bidirectional NK/DC interactions promote CD4 expression on NK cells, DC maturation, and HIV infection. Virology 2012; 433:203-15. [PMID: 22921314 DOI: 10.1016/j.virol.2012.06.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 06/15/2012] [Accepted: 06/22/2012] [Indexed: 11/17/2022]
Abstract
Interactions between natural killer (NK) and dendritic cells (DCs) are integral to immune response development, potentially leading to bidirectional NK/DC activation. We demonstrate that autologous NK/DC interactions induce CD4 expression on NK cells, influencing degranulation. Cell contact is required, with high NK:DC ratios and mature DCs most effectively inducing CD4 expression. CD4(+) NK cells, in turn, mediate DC maturation via contact-dependent and independent pathways, more effectively maturing DCs than CD4(-) NK cells. Bidirectional NK/DC interactions also impact HIV infection, as NK-matured DCs effectively deliver infectious HIV to T cells, via trans-infection. DC-induced CD4 expression also renders NK cells susceptible to HIV infection. Focusing on NK/DC interactions, DCs can transfer infectious virus and enhance HIV infection of CD4(+) NK cells, strongly suggesting that these interactions influence HIV pathogenesis. Findings provide new insight regarding NK/DC interactions, defining a mechanism by which cellular interactions in the absence of pathogens promote DC-mediated amplification of HIV infection.
Collapse
Affiliation(s)
- Alice Valentin-Torres
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | | | | |
Collapse
|
18
|
Marginal zone macrophages suppress innate and adaptive immunity to apoptotic cells in the spleen. Blood 2011; 117:5403-12. [PMID: 21444914 DOI: 10.1182/blood-2010-11-320028] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Marginal zone macrophages (MZMs) are a small subset of specialized splenic macrophages known to interact with apoptotic material entering the spleen from circulation. To evaluate whether MZMs regulate immunity to apoptotic material we depleted MZMs and assessed innate and adaptive immune responses to apoptotic cells administered systemically. MZM depletion altered the spatial localization of apoptotic cells, which accumulated in T-cell areas of the lymphoid follicles. MZM depletion also enhanced phagocytosis of apoptotic cells by red pulp (CD68(+)F4/80(+)) macrophages, which expressed increased CD86, MHCII, and CCR7. MZM depletion led to increased production of proinflammatory cytokines and enhanced lymphocyte responsiveness to apoptotic cell antigens. Furthermore, we found that MZM depletion accelerated autoimmune disease progression in mice genetically prone to systemic lupus erythematosus and caused significant mortality in wild-type mice repeatedly exposed to exogenous apoptotic thymocytes. These findings support the hypothesis that MZMs are central in the clearance of apoptotic cells to minimize the immunogenicity of autoantigens.
Collapse
|
19
|
da Costa TB, Sardinha LR, Larocca R, Peron JPS, Rizzo LV. Allogeneic apoptotic thymocyte-stimulated dendritic cells expand functional regulatory T cells. Immunology 2011; 133:123-32. [PMID: 21355864 DOI: 10.1111/j.1365-2567.2011.03420.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Dendritic cells (DCs) play an important role in the clearance of apoptotic cells. The removal of apoptotic cells leads to peripheral tolerance, although their role is still not clear. We show that the uptake of apoptotic thymocytes by DCs converts these cells into tolerogenic DCs resistant to maturation by lipopolysaccharide, modulating the production of interleukin-12 and up-regulating the expression of transforming growth factor-β(1) latency associated peptide. We also observed that DCs pulsed with apoptotic cells in the allogeneic context were more efficient in the expansion of regulatory T cells (Tregs), and that this expansion requires contact between DCs and the T cell. The Tregs sorted from in vitro culture suppressed the proliferation of splenocytes in vitro in a specific and non-specific manner. In the in vivo model, the transfer of CD4(+) CD25(-) cells to Nude mice induced autoimmunity, with cell infiltrate found in the stomach, colon, liver and kidneys. The co-transfer of CD4(+) CD25(-) and CD4(+) CD25(+) prevented the presence of cell infiltrates in several organs and increased the total cell count in lymph nodes. Our data indicate that apoptotic cells have an important role in peripheral tolerance via induction of tolerogenic DCs and CD4(+) CD25(+) Foxp3(+) cells that present regulatory functions.
Collapse
Affiliation(s)
- Thaís Boccia da Costa
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | | | | | | | | |
Collapse
|
20
|
Abstract
One of the ultimate goals in transplantation is to develop novel therapeutic methods for induction of donor-specific tolerance to reduce the side effects caused by the generalized immunosuppression associated to the currently used pharmacologic regimens. Interaction or phagocytosis of cells in early apoptosis exerts potent anti-inflammatory and immunosuppressive effects on antigen (Ag)-presenting cells (APC) like dendritic cells (DC) and macrophages. This observation led to the idea that apoptotic cell-based therapies could be employed to deliver donor-Ag in combination with regulatory signals to recipient’s APC as therapeutic approach to restrain the anti-donor response. This review describes the multiple mechanisms by which apoptotic cells down-modulate the immuno-stimulatory and pro-inflammatory functions of DC and macrophages, and the role of the interaction between apoptotic cells and APC in self-tolerance and in apoptotic cell-based therapies to prevent/treat allograft rejection and graft-versus-host disease in murine experimental systems and in humans. It also explores the role that in vivo-generated apoptotic cells could have in the beneficial effects of extracorporeal photopheresis, donor-specific transfusion, and tolerogenic DC-based therapies in transplantation.
Collapse
|
21
|
Kushwah R, Wu J, Oliver JR, Jiang G, Zhang J, Siminovitch KA, Hu J. Uptake of apoptotic DC converts immature DC into tolerogenic DC that induce differentiation of Foxp3+ Treg. Eur J Immunol 2010; 40:1022-35. [PMID: 20101618 DOI: 10.1002/eji.200939782] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
DC apoptosis has been observed in patients with cancer and sepsis, and defects in DC apoptosis have been implicated in the development of autoimmune diseases. However, the mechanisms of how DC apoptosis affects immune responses, are unclear. In this study, we showed that immature viable DC have the ability to uptake apoptotic DC as well as necrotic DC without it being recognized as an inflammatory event by immature viable DC. However, the specific uptake of apoptotic DC converted immature viable DC into tolerogenic DC, which were resistant to LPS-induced maturation. These tolerogenic DC secreted increased levels of TGF-beta1, which induced differentiation of naïve T cells into Foxp3(+) Treg. Furthermore, induction of Treg differentiation only occurred upon uptake of apoptotic DC and not apoptotic splenocytes by viable DC, indicating that it is specifically the uptake of apoptotic DC that gives viable immature DC the potential to induce Foxp3(+) Treg. Taken together, these findings identify uptake of apoptotic DC by viable immature DC as an immunologically tolerogenic event.
Collapse
Affiliation(s)
- Rahul Kushwah
- Physiology and Experimental Medicine Research Program, Hospital for Sick Children, Toronto, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
22
|
Kushwah R, Oliver JR, Zhang J, Siminovitch KA, Hu J. Apoptotic dendritic cells induce tolerance in mice through suppression of dendritic cell maturation and induction of antigen-specific regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:7104-18. [PMID: 19917707 DOI: 10.4049/jimmunol.0900824] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cell (DC) apoptosis has been shown to play a role in maintaining a balance between tolerance and immunity. However, the mechanisms of how DC apoptosis affects the immune response are unclear. We have shown that in vitro culture of apoptotic DCs with immature DCs, results in their uptake by immature DCs, which subsequently turn into tolerogenic DCs, which then secrete TGF-beta1 and induce Foxp3(+) regulatory T cells (T(regs)). In this study we looked at the effects of apoptotic DCs in vivo. Here we show that apoptotic DCs are taken up by viable DCs in vivo, which suppresses the ability of viable DCs to undergo maturation and subsequent migration to the lymph nodes in response to LPS. Additionally, delivery of apoptotic DCs to LPS inflamed lungs results in resolution of inflammation, which is mediated by the ability of apoptotic DCs to suppress response of viable DCs to LPS. Additionally, apoptotic DCs also induce TGF-beta1 secretion in the mediastinal lymph nodes, which results in expansion of Foxp3(+) T(regs). Most importantly, we show that delivery of apoptotic DCs followed by OVA in CFA to mice suppresses T cell response to OVA and instead induces de novo generation of OVA-specific T(regs). Furthermore, delivery of apoptotic DCs followed by OVA in CFA results in expansion of T(regs) in TCR transgenic (OT-II) mice. These findings demonstrate that apoptotic DCs are taken up by viable DCs in vivo, which promotes tolerance through suppression of DC maturation and induction of T(regs).
Collapse
Affiliation(s)
- Rahul Kushwah
- Physiology and Experimental Medicine Research Program, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | | | | | | | | |
Collapse
|
23
|
Chen Y, Khanna S, Goodyear CS, Park YB, Raz E, Thiel S, Grönwall C, Vas J, Boyle DL, Corr M, Kono DH, Silverman GJ. Regulation of dendritic cells and macrophages by an anti-apoptotic cell natural antibody that suppresses TLR responses and inhibits inflammatory arthritis. THE JOURNAL OF IMMUNOLOGY 2009; 183:1346-59. [PMID: 19564341 DOI: 10.4049/jimmunol.0900948] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Although natural Abs (NAbs) are present from birth, little is known about what drives their selection and whether they have housekeeping functions. The prototypic T15-NAb, first identified because of its protective role in infection, is representative of a special type of NAb response that specifically recognizes and forms complexes with apoptotic cells and which promotes cell-corpse engulfment by phagocytes. We now show that this T15-NAb IgM-mediated clearance process is dependent on the recruitment of C1q and mannose-binding lectin, which have known immune modulatory activities that also provide "eat me" signals for enhancing phagocytosis. Further investigation revealed that the addition of T15-NAb significantly suppressed in vitro LPS-induced TNF-alpha and IL-6 secretion by the macrophage-like cell line, RAW264.7, as well as TLR3-, TLR4-, TLR7-, and TLR9-induced maturation and secretion of a range of proinflammatory cytokines and chemokines by bone marrow-derived conventional dendritic cells. Significantly, high doses of this B-1 cell produced NAb also suppressed in vivo TLR-induced proinflammatory responses. Although infusions of apoptotic cells also suppressed such in vivo inflammatory responses and this effect was associated with the induction of high levels of IgM antiapoptotic cell Abs, apoptotic cell treatment was not effective at suppressing such TLR responses in B cell-deficient mice. Moreover, infusions of T15-NAb also efficiently inhibited both collagen-induced arthritis and anti-collagen II Ab-mediated arthritis. These studies identify and characterize a previously unknown regulatory circuit by which a NAb product of innate-like B cells aids homeostasis by control of fundamental inflammatory pathways.
Collapse
Affiliation(s)
- Yifang Chen
- Laboratory of B Cell Immunobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
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
|
25
|
Di Renzo M, Sbano P, De Aloe G, Pasqui AL, Rubegni P, Ghezzi A, Auteri A, Fimiani M. Extracorporeal photopheresis affects co-stimulatory molecule expression and interleukin-10 production by dendritic cells in graft-versus-host disease patients. Clin Exp Immunol 2008; 151:407-13. [PMID: 18234053 DOI: 10.1111/j.1365-2249.2007.03577.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Graft-versus-host disease (GVHD) is a major complication of allogeneic bone marrow transplantation. Extracorporeal photochemotherapy (ECP) has been introduced as an alternative treatment for GVHD refractory to conventional immunosuppressive treatment, although its mechanism of action is not yet clear. We investigated, in seven GVHD patients, the effects of ECP on dendritic cell maturation and cytokine production in an in vitro model that could mimic the potential in vivo effect of reinfusion of ECP-treated peripheral blood mononuclear cells. The model was based on co-culture of ECP-treated lymphocytes with monocyte-derived dendritic cells (DCs) of the same patient. We found that the co-culture of ECP-treated lymphocytes with immature DCs reduced CD54, CD40 and CD86 mean fluorescence intensity (MFI) significantly after lipopolysaccharide (LPS) stimulation, without affecting human leucocyte antigen D-related and CD80 MFI. In the same co-culture model, DCs produced increased amounts of interleukin (IL)-10 when co-cultured with ECP-treated lymphocytes and stimulated with LPS, while IL-12 and tumour necrosis factor-alpha production were not affected. These results suggest that reinfusion of large numbers of autologous apoptotic lymphocytes is significant for the therapeutic outcome of ECP through down-regulation of co-stimulatory molecules on DCs, inducing non-fully mature DCs with a low signal 2 and up-regulation of IL-10, which is an immunosuppressive cytokine.
Collapse
Affiliation(s)
- M Di Renzo
- Azienda Ospedaliera Universitaria Senese, University of Siena, Siena, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Behrens EM, Sriram U, Shivers DK, Gallucci M, Ma Z, Finkel TH, Gallucci S. Complement receptor 3 ligation of dendritic cells suppresses their stimulatory capacity. THE JOURNAL OF IMMUNOLOGY 2007; 178:6268-79. [PMID: 17475855 DOI: 10.4049/jimmunol.178.10.6268] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To activate T cells effectively, dendritic cells (DCs) must provide three separate signals, MHC-Ag, costimulatory molecules (such as CD80 and CD86), and proinflammatory cytokines (such as IL-12). These three signals are up-regulated in the presence of "danger signals" such as LPS or viral nucleic acids. Evidence suggests that DCs providing only the first two of these signals cannot successfully stimulate T cells. Apoptotic cells have been proposed to suppress DC immunogenicity through the ligation of apoptotic cell receptors. Complement receptor 3 (CR3) and CD36 have been suggested to be important in this process, although the mechanism by which this modulation occurs is still unclear. We demonstrate that ligation of CR3, but not CD36, directs DCs to increase surface MHC and costimulatory molecules, while suppressing inflammatory cytokine release. CR3 modulation of DCs does not require a type I IFN response, does not involve the specific regulation of the MyD88- or Toll/IL-1R domain-containing adaptor-inducing IFN-beta-dependent TLR signaling pathways, and occurs even in the absence of danger signals. The functional outcome of this process is poor Ag-specific stimulation of CD4 and CD8 T cells by CR3-ligated DCs both in naive response as well as upon subsequent challenge with normal DCs. We propose that CR3 provides a "nondanger" signal that suppresses the stimulatory capacity of DCs.
Collapse
Affiliation(s)
- Edward M Behrens
- Laboratory of Dendritic Cell Biology, Joseph Stokes, Jr. Research Institute, Children's Hospital of Philadelphia, and Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-4318, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
The termination of the apoptotic programme occurs in most cases via recognition and clearance by phagocytes, especially the professional phagocytes, such as macrophages and immature dendritic cells. Engulfed cells do not simply disappear from the midst of living tissues. The fine-defined presentation of yielded self-antigens to T cells is a central event in the induction or the maintenance of the peripheral immune tolerance to self. Conversely, abnormality in this pathway may contribute to the pathogenesis of systemic and organ-specific autoimmune diseases. We herein reviewed the relationship between phagocytosis of apoptotic cells and immune regulation, especially the effects of engulfed apoptotic cells on immune tolerance and autoimmune diseases.
Collapse
Affiliation(s)
- G Liu
- Transplantation Biology Research Division, State Key Laboratory of Biomembrane and Membrane Biotechnology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | | | | | | |
Collapse
|
28
|
Gupta S, Agrawal A, Agrawal S, Su H, Gollapudi S. A paradox of immunodeficiency and inflammation in human aging: lessons learned from apoptosis. Immun Ageing 2006; 3:5. [PMID: 16712718 PMCID: PMC1475884 DOI: 10.1186/1742-4933-3-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2005] [Accepted: 05/19/2006] [Indexed: 01/05/2023]
Abstract
Aging is associated with a paradox of immunodeficiency and inflammation (an evidence of hyperactive immune system). Apoptosis is associated with cellular depletion and suppression of inflammatory response. In this brief review, we will present evidence for the role of increased apoptosis in immunodeficiency and paradoxical increased inflammation associated with human aging. In particular, a role of apoptotic cells in failure to generate anti-inflammatory responses and directly activating inflammatory responses will be discussed.
Collapse
Affiliation(s)
- Sudhir Gupta
- Laboratories of Cellular and Molecular Immunology, Division of Basic and Clinical Immunology, University of California, Irvine, California 92697, USA
| | - Anshu Agrawal
- Laboratories of Cellular and Molecular Immunology, Division of Basic and Clinical Immunology, University of California, Irvine, California 92697, USA
| | - Sudhanshu Agrawal
- Laboratories of Cellular and Molecular Immunology, Division of Basic and Clinical Immunology, University of California, Irvine, California 92697, USA
| | - Houfen Su
- Laboratories of Cellular and Molecular Immunology, Division of Basic and Clinical Immunology, University of California, Irvine, California 92697, USA
| | - Sastry Gollapudi
- Laboratories of Cellular and Molecular Immunology, Division of Basic and Clinical Immunology, University of California, Irvine, California 92697, USA
| |
Collapse
|
29
|
Iyoda T, Nagata K, Akashi M, Kobayashi Y. Neutrophils Accelerate Macrophage-Mediated Digestion of Apoptotic Cells In Vivo as Well as In Vitro. THE JOURNAL OF IMMUNOLOGY 2005; 175:3475-83. [PMID: 16148089 DOI: 10.4049/jimmunol.175.6.3475] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It is generally believed that the clearance of apoptotic cells does not lead to inflammation. In contrast, we previously found that injection of apoptotic cells into the peritoneal cavity induced the expression of an inflammatory chemokine, MIP-2, and infiltration of neutrophils, and that anti-MIP-2 Abs suppressed the infiltration significantly. Because our previous study showed that whole-body x-irradiation caused neutrophil infiltration into the thymus along with T cell apoptosis, we examined the role of neutrophils in apoptotic cell clearance. Neutrophil infiltration reached a peak 12 h after irradiation with 1 Gy of x-rays. Immunohistological analysis revealed that apoptotic cells disappeared dramatically from 10.5 to 12 h after x-irradiation. As neutrophils moved from an inner area of the cortex to the periphery, apoptotic cells disappeared concomitantly. Either anti-MIP-2 or anti-CXCR2 Abs suppressed neutrophil infiltration significantly, and the suppression of neutrophil infiltration by anti-MIP-2 Abs delayed the disappearance of apoptotic cells. Moreover, macrophage-mediated digestion of apoptotic thymocytes was accelerated in vitro on coculturing with neutrophils, even if neutrophils were separated from macrophages. These results suggest that neutrophils are recruited to the thymus mainly by MIP-2 after whole-body x-irradiation and that such neutrophils may not induce inflammation but rather accelerate complete digestion of apoptotic cells by macrophages.
Collapse
Affiliation(s)
- Takuya Iyoda
- Division of Molecular Medicine, Department of Biomolecular Science, Faculty of Science, Toho University, Funabashi, Japan
| | | | | | | |
Collapse
|
30
|
Abstract
The paradigm of tolerogenic/immature versus inflammatory/mature dendritic cells has dominated the recent literature regarding the role of these antigen-presenting cells in mediating immune homeostasis or self-tolerance and response to pathogens, respectively. This issue is further complicated by the identification of distinct subtypes of dendritic cells that exhibit different antigen-presenting cell effector functions. The discovery of pathogen-associated molecular patterns and toll-like receptors provides the mechanistic basis for dendritic cell recognition of specific pathogens and induction of appropriate innate and adaptive immune responses. Only recently has insight been gained into how dendritic cells contribute to establishing and/or maintaining immunological tolerance to self. Soluble and cellular mediators have been reported to effectively regulate the function of dendritic cells by inducing several outcomes ranging from non-inflammatory dendritic cells that lack the ability to induce T lymphocyte activation to dendritic cells that actively suppress T lymphocyte responses. A thorough discussion of these stimuli and their outcomes is essential to understanding the potential for modulating dendritic cell function in the treatment of inflammatory disease conditions.
Collapse
Affiliation(s)
- Mark A Wallet
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, CB7290, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
31
|
Alexis NE, Lay JC, Almond M, Bromberg PA, Patel DD, Peden DB. Acute LPS inhalation in healthy volunteers induces dendritic cell maturation in vivo. J Allergy Clin Immunol 2005; 115:345-50. [PMID: 15696093 DOI: 10.1016/j.jaci.2004.11.040] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND We have been studying the innate immune response of airways cells of healthy human volunteers to inhaled LPS, a Toll-like receptor 4 (TLR4) ligand, and have shown that macrophage phagocytic capacity is blunted. OBJECTIVE Because a primary feature of dendritic cell (DC) maturation is a loss of phagocytic capacity, we sought to determine whether acute LPS inhalation in healthy volunteers promotes DC maturation in vivo. METHODS Phagocytosis (IgG-opsonized zymosan particles) and cell-surface phenotypes were analyzed by flow cytometry of induced sputum cells obtained before and 6 hours after Clinical Center Reference Endotoxin (CCRE; 20,000 EU) inhalation in 9 healthy volunteers. RESULTS Neutrophils were elevated in the airways after CCRE inhalation (67% +/- 6% vs 37% +/- 6%; P < .05). Phagocytosis (monocytes, macrophages) was blunted (73%, 46%; P < .05) and negatively correlated with PMN influx ( R = -0.73; P < .05) after CCRE inhalation. GM-CSF and IL-1beta, potent DC maturation agents, were elevated after versus before CCRE inhalation (217 pg/mL +/- 103 pg/mL vs 722 pg/mL +/- 202 pg/mL; 83 pg/mL +/- 24 pg/mL vs 148 pg/mL +/- 37 pg/mL, respectively; P < .05). Markers of DC maturation (CD80, CD86, HLA-DR) were upregulated on monocytes and macrophages ( P < .05), and discrete populations of mature DC were observed ( P < .05) after CCRE inhalation. CONCLUSION Inhaled LPS, directly through TLR4 stimulation of immature DC and/or indirectly through stimulation of GM-CSF and IL-1beta, induces pulmonary DC maturation in vivo . Inhaled LPS may enhance allergic airways responses to air pollution through its ability to induce DC maturation.
Collapse
Affiliation(s)
- Neil E Alexis
- Center for Environmental Biology, Asthma, and Lung Biology, University of North Carolina at Chapel Hill, NC 27599, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Chen X, Doffek K, Sugg SL, Shilyansky J. Phosphatidylserine regulates the maturation of human dendritic cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:2985-94. [PMID: 15322157 DOI: 10.4049/jimmunol.173.5.2985] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Phosphatidylserine (PS), which is exposed on the surface of apoptotic cells, has been implicated in immune regulation. However, the effects of PS on the maturation and function of dendritic cells (DCs), which play a central role in both immune activation and regulation, have not been described. Large unilamellar liposomes containing PS or phosphatidylcholine were used to model the plasma membrane phospholipid composition of apoptotic and live cells, respectively. PS liposomes inhibited the up-regulation of HLA-ABC, HLA-DR, CD80, CD86, CD40, and CD83, as well as the production of IL-12p70 by human DCs in response to LPS. PS did not affect DC viability directly but predisposed DCs to apoptosis in response to LPS. DCs exposed to PS had diminished capacity to stimulate allogeneic T cell proliferation and to activate IFN-gamma-producing CD4(+) T cells. Exogenous IL-12 restored IFN-gamma production by CD4(+) T cells. Furthermore, activated CTLs proliferated poorly to cognate Ag presented by DCs exposed to PS. Our findings suggest that PS exposure provides a sufficient signal to inhibit DC maturation and to modulate adaptive immune responses.
Collapse
Affiliation(s)
- Xiao Chen
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee 53226, USA
| | | | | | | |
Collapse
|