1
|
Wang S, Zhang N, Shi G, Liu X, Zhou Y, Yang H. Genome-wide chromatin accessibility and transcriptome analysis reveal the up-regulation of immunosuppressive genes in macrophages under simulated microgravity. LIFE SCIENCES IN SPACE RESEARCH 2025; 45:143-150. [PMID: 40280635 DOI: 10.1016/j.lssr.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/07/2025] [Accepted: 03/11/2025] [Indexed: 04/29/2025]
Abstract
Long-term space missions are of growing research interest because of the space exploration. However, plenty of works focused on the impaired immune response, less attention has been paid to the activation of immunosuppressive or anti-inflammatory function. The molecular mechanism of immune disorder induced by microgravity still needs investigation. Here, we used a random positioning machine to generate a simulated microgravity environment and evaluated its effects on mouse RAW 264.7 macrophage cell line. We used ATAC-seq and RNA-seq for revealing the mechanism at chromatin level and gene level. From ATAC-seq, we obtained an average of 75,700,675 paired-end clean reads for each library and the mapping rates averaged at 96.8 %. The number of differential accessible regions were 510 for increased peaks, 638 for decreased peaks. From RNA-seq, we obtained 278 differentially expressed genes, of which 104 were down-regulated and 174 were up-regulated genes. Through ATAC-seq and RNA-seq multi-omics analysis, we identified a group of 17 genes. Then we chose 6 up-regulated genes (CD83, CEBPD, CXCR5, DUSP6, SEMA4B, TNFRSF22) that related to immunosuppressive function for further confirmation. The qRT-PCR results were consistent with sequencing results, which indicated that simulated microgravity leads to the up-regulated expression of immunosuppressive genes of macrophages. Taken together, our results offered novel insights for understanding the brief principles and mechanisms of simulated microgravity induced immune dysfunction to macrophage.
Collapse
Affiliation(s)
- Sufang Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Center of Special Environmental Biomechanics & Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Nu Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Center of Special Environmental Biomechanics & Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Guolin Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Center of Special Environmental Biomechanics & Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Xiru Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Center of Special Environmental Biomechanics & Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Yidan Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Center of Special Environmental Biomechanics & Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Center of Special Environmental Biomechanics & Biomedical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment in Special Environment, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
2
|
Harris AR, Pichardo CM, Franklin J, Liu H, Wooten W, Panigrahi G, Lawrence WR, Pichardo MS, Jenkins BD, Dorsey TH, Ioffe OB, Yfantis HG, Agurs-Collins T, Ambs S. Multilevel Stressors and Systemic and Tumor Immunity in Black and White Women With Breast Cancer. JAMA Netw Open 2025; 8:e2459754. [PMID: 39951265 PMCID: PMC11829235 DOI: 10.1001/jamanetworkopen.2024.59754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/05/2024] [Indexed: 02/17/2025] Open
Abstract
Importance The mechanisms through which chronic stressors may be associated with tumor biologic characteristics, immune response, and health disparities remain insufficiently understood. Objective To investigate the proteomic, transcriptomic, and genomic effects associated with multilevel chronic stressors (perceived stress, perceived inadequate social support, perceived racial and ethnic discrimination, and neighborhood deprivation) in Black and White women with breast cancer. Design, Setting, and Participants This cross-sectional study was conducted from February 28, 2012, to September 5, 2023, in which blood samples, breast tumors, and adjacent noncancerous tissues were collected from women with breast cancer. Participants, recruited at 2 Baltimore, Maryland, hospitals, completed demographic and psychosocial questionnaires. Data analysis was conducted from September 2023 to April 2024. Exposures Perceived stress, perceived social support, perceived racial and ethnic discrimination, and the 2010 census tract-level neighborhood deprivation index, in which scores range from -2.51 to 6.77, with higher scores indicating greater deprivation. Main Outcomes and Measures The main outcomes included levels of 92 circulating immune-oncologic markers and associated biologic pathways, tumor immune cell profiles, breast tissue gene expression, and tumor mutational burden. Data were analyzed using covariate-adjusted linear regression modeling for continuous outcomes with effect estimates presented as β values with 95% CIs. Results The study included 121 women with breast cancer (mean [SD] age, 56.27 [12.62] years), of whom 56 (46.3%) were Black, and 65 (53.7%) were White. The analytic subsample sizes included 117 blood samples, 48 breast tumors, and 41 adjacent noncancerous tissues. Levels of perceived stress and social support were comparable by race, while Black women resided in more socioeconomically deprived neighborhoods (mean [SD] neighborhood deprivation index, 2.28 [2.30] for Black women compared with -0.22 [2.01] for White women). Greater perceived social support was associated with more favorable immune-stimulatory changes (eg, increased serum IL-5 [β, 0.06 (95% CI, 0.02-0.10); P = .003] and activated natural killer cells in noncancerous breast tissue of Black women [β, 0.11 (95% CI, 0.04-0.17); P = .002). Higher levels of perceived stress, exposure to discrimination, and neighborhood deprivation were associated with systemic inflammation (eg, serum IL-6 with both perceived stress [β, 0.04 (95% CI, 0.01-0.07); P = .006] and discrimination [β, 0.69 (95% CI, 0.15-1.23); P = .01]); deleterious immune cell profiles (eg, tumor-associated M2 macrophages with discrimination [β, 0.82 (95% CI, 0.14-1.51); P = .02]); and aggressive tumor biologic characteristics. Race-stratified analyses uncovered distinct immunologic features in Black women associated with stressors, including chemotaxis with stress (β, 0.28 [95% CI, 0.001-0.56]; P = .049) and immune suppression with stress (β, 0.37 [95% CI, -0.002 to 0.75]; P = .05) at the systemic level and increased tumor-associated myeloid cells (monocytes and M1 and M2 macrophages) at the tissue level. Perceived stress was associated with elevated tumor mutational burden (β, 0.02 [95% CI, 0.01-0.04]; P = .04). Conclusions and Relevance The findings of this cross-sectional study of Black and White women with breast cancer suggest that perceived stress, perceived inadequate social support, perceived racial and ethnic discrimination, and neighborhood deprivation were associated with deleterious alterations to the systemic and tumor immune environment, particularly for Black women. Understanding biology as a possible mediator of cancer health disparities may inform prevention and public health interventions.
Collapse
Affiliation(s)
- Alexandra R. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Catherine M. Pichardo
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Jamirra Franklin
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - William Wooten
- University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center Biostatistics Shared Service, Baltimore
| | - Gatikrushna Panigrahi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Wayne R. Lawrence
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | | | - Brittany D. Jenkins
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Tiffany H. Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Olga B. Ioffe
- Department of Pathology, University of Maryland School of Medicine, Baltimore
| | - Harry G. Yfantis
- Pathology and Laboratory Medicine, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| | - Tanya Agurs-Collins
- Division of Cancer Control and Population Sciences, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
3
|
Tzaban S, Stern O, Zisman E, Eisenberg G, Klein S, Frankenburg S, Lotem M. Alternative splicing of modulatory immune receptors in T lymphocytes: a newly identified and targetable mechanism for anticancer immunotherapy. Front Immunol 2025; 15:1490035. [PMID: 39845971 PMCID: PMC11752881 DOI: 10.3389/fimmu.2024.1490035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Abstract
Alternative splicing (AS) is a mechanism that generates translational diversity within a genome. Equally important is the dynamic adaptability of the splicing machinery, which can give preference to one isoform over others encoded by a single gene. These isoform preferences change in response to the cell's state and function. Particularly significant is the impact of physiological alternative splicing in T lymphocytes, where specific isoforms can enhance or reduce the cells' reactivity to stimuli. This process makes splicing isoforms defining features of cell states, exemplified by CD45 splice isoforms, which characterize the transition from naïve to memory states. Two developments have accelerated the use of AS dynamics for therapeutic interventions: advancements in long-read RNA sequencing and progress in nucleic acid chemical modifications. Improved oligonucleotide stability has enabled their use in directing splicing to specific sites or modifying sequences to enhance or silence particular splicing events. This review highlights immune regulatory splicing patterns with potential significance for enhancing anticancer immunotherapy.
Collapse
Affiliation(s)
- Shay Tzaban
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ori Stern
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Elad Zisman
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Galit Eisenberg
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Center for Melanoma and Cancer Immunotherapy, Sharett Institute of Oncology, Jerusalem, Israel
| | - Shiri Klein
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Center for Melanoma and Cancer Immunotherapy, Sharett Institute of Oncology, Jerusalem, Israel
| | - Shoshana Frankenburg
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Lotem
- The Lautenberg Center for Immunology and Cancer Research, The Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Center for Melanoma and Cancer Immunotherapy, Sharett Institute of Oncology, Jerusalem, Israel
- Hadassah Cancer Research Institute, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
4
|
Krupa P, Wein H, Zemmrich LS, Zygmunt M, Muzzio DO. Pregnancy-related factors induce immune tolerance through regulation of sCD83 release. Front Immunol 2024; 15:1452879. [PMID: 39328416 PMCID: PMC11424458 DOI: 10.3389/fimmu.2024.1452879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
A well-balanced maternal immune system is crucial to maintain fetal tolerance in case of infections during pregnancy. Immune adaptations include an increased secretion of soluble mediators to protect the semi-allogeneic fetus from excessive pro-inflammatory response. B lymphocytes acquire a higher capacity to express CD83 and secrete soluble CD83 (sCD83) upon exposure to bacteria-derived components such as LPS. CD83 possesses immune modulatory functions and shows a promising therapeutic potential against inflammatory conditions. The administration of sCD83 to pregnant mice reduces LPS-induced abortion rates. The increased CD83 expression by endometrial B cells as compared to peripheral blood B cells suggests its modulatory role in the fetal tolerance, especially in the context of infection. We postulate that in pregnancy, CD83 expression and release is controlled by pregnancy-related hormones. The intra- and extracellular expression of CD83 in leukocytes from peripheral blood or decidua basalis and parietalis at term were analyzed by flow cytometry. After treatment with pregnancy-related hormones and LPS, ELISA and qPCR were performed to study sCD83 release and CD83 gene expression, respectively. Cleavage prediction analysis was used to find potential proteases targeting CD83. Expression of selected proteases was analyzed by ELISA. Higher levels of CD83 were found in CD11c+ dendritic cells, CD3+ T cells and CD19+ B cells from decidua basalis and decidua parietalis after LPS-stimulation in vitro. An increase of intracellular expression of CD83 was also detected in CD19+ B cells from both compartments. Stimulated B cells displayed significantly higher percentages of CD83+ cells than dendritic cells and T cells from decidua basalis and peripheral blood. Treatment of B lymphocytes with pregnancy-related molecules (E2, P4, TGF-β1 and hCG) enhanced the LPS-mediated increase of CD83 expression, while dexamethasone led to a reduction. Similarly, the release of sCD83 was increased under TGF-β1 treatment but decreased upon dexamethasone stimulation. Finally, we found that the hormonal regulation of CD83 expression is likely a result from a balance between gene transcription from CD83 and the modulation of the metalloproteinase MMP-7. Thus, data supports and complements our previous murine studies on hormonal regulation of CD83 expression, reinforcing its immunomodulatory relevance in anti-bacterial responses during pregnancy.
Collapse
Affiliation(s)
| | | | | | | | - Damián Oscar Muzzio
- Research Laboratory, Department of Obstetrics and Gynecology, University Medicine
Greifswald, Greifswald, Germany
| |
Collapse
|
5
|
Li LH, Hsu DZ, Chandrasekaran VRM, Liu MY. Inhibiting CD44-ICD Attenuates LPS-Induced Initiation of Hepatic Inflammation in Septic Mice. Int J Mol Sci 2024; 25:8907. [PMID: 39201593 PMCID: PMC11354311 DOI: 10.3390/ijms25168907] [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: 07/19/2024] [Revised: 08/10/2024] [Accepted: 08/13/2024] [Indexed: 09/02/2024] Open
Abstract
Sepsis is a severe condition induced by microbial infection. It elicits a systemic inflammatory response, leading to multi-organ failure, and the liver, as a scavenger, plays a significant role in this process. Controlling hepatic inflammation and maintaining liver function is crucial in managing sepsis. CD44-ICD, as a CD44 signal transductor, is involved in multiple inflammatory responses. However, the role of CD44-ICD in lipopolysaccharide (LPS)-induced hepatic inflammation has not been investigated. Therefore, we aimed to examine whether CD44-ICD initiates hepatic inflammation in septic mice. We induced hepatic inflammation in mice by administering LPS. DAPT, a CD44-ICD inhibitor, was given to mice or Chang cells 30 min or 1 h before LPS administration (10 mg/kg, i.p., or 100 ng/mL, respectively). Inhibition of CD44-ICD decreased the level of aspartate aminotransferase (AST), alanine aminotransferase (ALT), hepatic necrosis, inflammatory cell infiltration, interleukin (IL)-1β, inducible NO synthase (iNOS), nitric oxide (NO) production, nuclear factor (NF)κB signaling pathway proteins, and CD44 expression in mice. CD44-ICD inhibition also decreased IL-1β and CD44 expression levels in Chang cells. CD44-ICD may be a primary regulatory function in CD44-associated LPS-induced initiation of hepatic inflammation in mice.
Collapse
Affiliation(s)
| | | | - Victor Raj Mohan Chandrasekaran
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan
| | - Ming-Yie Liu
- Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, Tainan 70428, Taiwan
| |
Collapse
|
6
|
Münter R, Bak M, Thomsen ME, Parhamifar L, Stensballe A, Simonsen JB, Kristensen K, Andresen TL. Deciphering the monocyte-targeting mechanisms of PEGylated cationic liposomes by investigating the biomolecular corona. Int J Pharm 2024; 657:124129. [PMID: 38621615 DOI: 10.1016/j.ijpharm.2024.124129] [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: 01/02/2024] [Revised: 04/04/2024] [Accepted: 04/13/2024] [Indexed: 04/17/2024]
Abstract
Cationic liposomes specifically target monocytes in blood, rendering them promising drug-delivery tools for cancer immunotherapy, vaccines, and therapies for monocytic leukaemia. The mechanism behind this monocyte targeting ability is, however, not understood, but may involve plasma proteins adsorbed on the liposomal surfaces. To shed light on this, we investigated the biomolecular corona of three different types of PEGylated cationic liposomes, finding all of them to adsorb hyaluronan-associated proteins and proteoglycans upon incubation in human blood plasma. This prompted us to study the role of the TLR4 co-receptors CD44 and CD14, both involved in signalling and uptake pathways of proteoglycans and glycosaminoglycans. We found that separate inhibition of each of these receptors hampered the monocyte uptake of the liposomes in whole human blood. Based on clues from the biomolecular corona, we have thus identified two receptors involved in the targeting and uptake of cationic liposomes in monocytes, in turn suggesting that certain proteoglycans and glycosaminoglycans may serve as monocyte-targeting opsonins. This mechanistic knowledge may pave the way for rational design of future monocyte-targeting drug-delivery platforms.
Collapse
Affiliation(s)
- Rasmus Münter
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Martin Bak
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Mikkel E Thomsen
- Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark
| | - Ladan Parhamifar
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, 9260 Gistrup, Denmark; Clinical Cancer Center, Aalborg University Hospital, 9000 Aalborg, Denmark
| | - Jens B Simonsen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Kasper Kristensen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| | - Thomas L Andresen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
7
|
Maggiorani D, Le O, Lisi V, Landais S, Moquin-Beaudry G, Lavallée VP, Decaluwe H, Beauséjour C. Senescence drives immunotherapy resistance by inducing an immunosuppressive tumor microenvironment. Nat Commun 2024; 15:2435. [PMID: 38499573 PMCID: PMC10948808 DOI: 10.1038/s41467-024-46769-9] [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: 07/20/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
The potential of immune checkpoint inhibitors (ICI) may be limited in situations where immune cell fitness is impaired. Here, we show that the efficacy of cancer immunotherapies is compromised by the accumulation of senescent cells in mice and in the context of therapy-induced senescence (TIS). Resistance to immunotherapy is associated with a decrease in the accumulation and activation of CD8 T cells within tumors. Elimination of senescent cells restores immune homeostasis within the tumor micro-environment (TME) and increases mice survival in response to immunotherapy. Using single-cell transcriptomic analysis, we observe that the injection of ABT263 (Navitoclax) reverses the exacerbated immunosuppressive profile of myeloid cells in the TME. Elimination of these myeloid cells also restores CD8 T cell proliferation in vitro and abrogates immunotherapy resistance in vivo. Overall, our study suggests that the use of senolytic drugs before ICI may constitute a pharmacological approach to improve the effectiveness of cancer immunotherapies.
Collapse
Affiliation(s)
- Damien Maggiorani
- Centre de recherche du CHU Sainte-Justine, Montréal, QC, Canada
- Département de pharmacologie et physiologie (Université de Montréal, Montréal, QC, Canada
| | - Oanh Le
- Centre de recherche du CHU Sainte-Justine, Montréal, QC, Canada
| | - Véronique Lisi
- Centre de recherche du CHU Sainte-Justine, Montréal, QC, Canada
| | | | | | - Vincent Philippe Lavallée
- Centre de recherche du CHU Sainte-Justine, Montréal, QC, Canada
- Département de pédiatrie (Université de Montréal, Montréal, QC, Canada
| | - Hélène Decaluwe
- Centre de recherche du CHU Sainte-Justine, Montréal, QC, Canada
- Département de pédiatrie (Université de Montréal, Montréal, QC, Canada
- Département de microbiologie, immunologie et infectiologie (Université de Montréal, Montréal, QC, Canada
| | - Christian Beauséjour
- Centre de recherche du CHU Sainte-Justine, Montréal, QC, Canada.
- Département de pharmacologie et physiologie (Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
8
|
Peckert-Maier K, Wild AB, Sprißler L, Fuchs M, Beck P, Auger JP, Sinner P, Strack A, Mühl-Zürbes P, Ramadan N, Kunz M, Krönke G, Stich L, Steinkasserer A, Royzman D. Soluble CD83 modulates human-monocyte-derived macrophages toward alternative phenotype, function, and metabolism. Front Immunol 2023; 14:1293828. [PMID: 38162675 PMCID: PMC10755915 DOI: 10.3389/fimmu.2023.1293828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024] Open
Abstract
Alterations in macrophage (Mφ) polarization, function, and metabolic signature can foster development of chronic diseases, such as autoimmunity or fibrotic tissue remodeling. Thus, identification of novel therapeutic agents that modulate human Mφ biology is crucial for treatment of such conditions. Herein, we demonstrate that the soluble CD83 (sCD83) protein induces pro-resolving features in human monocyte-derived Mφ biology. We show that sCD83 strikingly increases the expression of inhibitory molecules including ILT-2 (immunoglobulin-like transcript 2), ILT-4, ILT-5, and CD163, whereas activation markers, such as MHC-II and MSR-1, were significantly downregulated. This goes along with a decreased capacity to stimulate alloreactive T cells in mixed lymphocyte reaction (MLR) assays. Bulk RNA sequencing and pathway analyses revealed that sCD83 downregulates pathways associated with pro-inflammatory, classically activated Mφ (CAM) differentiation including HIF-1A, IL-6, and cytokine storm, whereas pathways related to alternative Mφ activation and liver X receptor were significantly induced. By using the LXR pathway antagonist GSK2033, we show that transcription of specific genes (e.g., PPARG, ABCA1, ABCG1, CD36) induced by sCD83 is dependent on LXR activation. In summary, we herein reveal for the first time mechanistic insights into the modulation of human Mφ biology by sCD83, which is a further crucial preclinical study for the establishment of sCD83 as a new therapeutical agent to treat inflammatory conditions.
Collapse
Affiliation(s)
- Katrin Peckert-Maier
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Andreas B. Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Laura Sprißler
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Maximilian Fuchs
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Philipp Beck
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Jean-Philippe Auger
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Pia Sinner
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Astrid Strack
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Ntilek Ramadan
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Meik Kunz
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
- Chair of Medical Informatics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| | - Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich– Alexander Universität Erlangen–Nürnberg, Erlangen, Germany
| |
Collapse
|
9
|
Zamora-Pineda J, Kalinina O, Sperling AI, Knight KL. Mechanism of TLR4-Mediated Anti-Inflammatory Response Induced by Exopolysaccharide from the Probiotic Bacillus subtilis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1232-1239. [PMID: 37672039 DOI: 10.4049/jimmunol.2200855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 08/02/2023] [Indexed: 09/07/2023]
Abstract
Intestinal inflammatory diseases affect millions of people worldwide, and one class of drugs showing promise toward treatment of several inflammatory diseases is probiotics. Numerous studies have been performed using probiotics to prevent and treat intestinal inflammatory diseases. Most of these studies used intact bacteria, and neither the active molecule nor the molecular mechanisms by which they affect immune responses are known. We have shown that the probiotic Bacillus subtilis is anti-inflammatory and can protect mice from acute colitis induced by the enteric pathogen Citrobacter rodentium. We identified and purified the active molecule, exopolysaccharide (EPS), and showed that it protects mice from C. rodentium-induced colitis by inducing anti-inflammatory M2 macrophages or inhibitory dendritic cells (DCs), both of which inhibit excessive T cell responses. We showed previously that EPS affects macrophages and DCs in a TLR4-dependent manner, and in the current study we asked how EPS induces these anti-inflammatory cells and how they function to inhibit T cells. By investigating the signaling downstream of TLR4 that leads to acquisition of inhibitory properties of macrophages and DCs, we found that EPS induces expression of the inhibitory molecule IDO in bone marrow-derived DCs, and that inhibition of T cell proliferation by IDO-expressing bone marrow-derived DCs utilizes the kynurenine/aryl hydrocarbon receptor circuit. Furthermore, unlike LPS, EPS does not induce inflammatory cytokines upon injection in vivo, directly demonstrating different outcomes induced by two different TLR4 agonists.
Collapse
Affiliation(s)
- Jesus Zamora-Pineda
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Olga Kalinina
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| | - Anne I Sperling
- Pulmonary and Critical Care Division, Department of Medicine, University of Virginia, Charlottesville, VA
| | - Katherine L Knight
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, IL
| |
Collapse
|
10
|
Zizzo G, Cohen PL. Editorial: The key role of Mer receptor tyrosine kinase: where inflammation ends and fibrosis begins. Front Immunol 2023; 14:1251577. [PMID: 37529052 PMCID: PMC10390068 DOI: 10.3389/fimmu.2023.1251577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 07/10/2023] [Indexed: 08/03/2023] Open
Affiliation(s)
- Gaetano Zizzo
- Temple Autoimmunity Center, Temple University, Philadelphia, PA, United States
- Unit of Rheumatology, Department of Internal Medicine, Azienda Socio-Sanitaria Territoriale (ASST) Ovest Milanese, Milan, Italy
| | - Philip L. Cohen
- Temple Autoimmunity Center, Temple University, Philadelphia, PA, United States
- Section of Rheumatology, Department of Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
11
|
Porcine Reproductive and Respiratory Syndrome Virus Modulates the Switch of Macrophage Polarization from M1 to M2 by Upregulating MoDC-Released sCD83. Viruses 2023; 15:v15030773. [PMID: 36992481 PMCID: PMC10054646 DOI: 10.3390/v15030773] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/16/2023] [Accepted: 03/16/2023] [Indexed: 03/22/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV), the most economically important infectious disease of pigs, elicits poor innate and adaptive immune responses. Soluble CD83 (sCD83), a secretion from various immune cell populations, especially MoDCs, is involved in negatively regulating the immune response. We speculate sCD83 may be a critical factor in the process of PRRSV-coordinated macrophage polarization. In this study, we found that PAMs co-cultured with PRRSV-infected MoDCs inhibited the M1 macrophage while enhancing the M2 macrophage. This was accompanied by a decrease in the pro-inflammatory cytokine TNF-α and iNOS and an increase in the anti-inflammatory cytokine IL-10 and Arg1. Meanwhile, sCD83 incubation causes the same specific effects lead to a switch in macrophage from M1 to M2. Neutralization of sCD83 removes the inhibitory effects of PRRSV on PAMs. Using reverse genetics, we generated recombinant PRRSVs with mutations in N protein, nsp1α, and nsp10 (knockout sCD83-concerned key amino acid site). Four mutant viruses lost the suppression of M1 macrophage markers, in contrast to the restriction of the upregulation of M2 macrophage markers. These findings suggest that PRRSV modulates the switch of macrophage polarization from M1 to M2 by upregulating the MoDC-induced secretion of CD83, providing new insights into the mechanism by which PRRSV regulates host immunity.
Collapse
|
12
|
Wu Z, Yoshikawa T, Inoue S, Ito Y, Kasuya H, Nakashima T, Zhang H, Kotaka S, Hosoda W, Suzuki S, Kagoya Y. CD83 expression characterizes precursor exhausted T cell population. Commun Biol 2023; 6:258. [PMID: 36906640 PMCID: PMC10008643 DOI: 10.1038/s42003-023-04631-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 02/27/2023] [Indexed: 03/13/2023] Open
Abstract
T cell exhaustion is a main obstacle against effective cancer immunotherapy. Exhausted T cells include a subpopulation that maintains proliferative capacity, referred to as precursor exhausted T cells (TPEX). While functionally distinct and important for antitumor immunity, TPEX possess some overlapping phenotypic features with the other T-cell subsets within the heterogeneous tumor-infiltrating T-lymphocytes (TIL). Here we explore surface marker profiles unique to TPEX using the tumor models treated by chimeric antigen receptor (CAR)-engineered T cells. We find that CD83 is predominantly expressed in the CCR7+PD1+ intratumoral CAR-T cells compared with the CCR7-PD1+ (terminally differentiated) and CAR-negative (bystander) T cells. The CD83+CCR7+ CAR-T cells exhibit superior antigen-induced proliferation and IL-2 production compared with the CD83- T cells. Moreover, we confirm selective expression of CD83 in the CCR7+PD1+ T-cell population in primary TIL samples. Our findings identify CD83 as a marker to discriminate TPEX from terminally exhausted and bystander TIL.
Collapse
Affiliation(s)
- Zhiwen Wu
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Toshiaki Yoshikawa
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Inoue
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yusuke Ito
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Hitomi Kasuya
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Takahiro Nakashima
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Haosong Zhang
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Saki Kotaka
- Department of Gynecologic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Waki Hosoda
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Shiro Suzuki
- Department of Gynecologic Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Yuki Kagoya
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan.
- Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan.
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
13
|
Riaz B, Islam SMS, Ryu HM, Sohn S. CD83 Regulates the Immune Responses in Inflammatory Disorders. Int J Mol Sci 2023; 24:ijms24032831. [PMID: 36769151 PMCID: PMC9917562 DOI: 10.3390/ijms24032831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Activating the immune system plays an important role in maintaining physiological homeostasis and defending the body against harmful infections. However, abnormalities in the immune response can lead to various immunopathological responses and severe inflammation. The activation of dendritic cells (DCs) can influence immunological responses by promoting the differentiation of T cells into various functional subtypes crucial for the eradication of pathogens. CD83 is a molecule known to be expressed on mature DCs, activated B cells, and T cells. Two isotypes of CD83, a membrane-bound form and a soluble form, are subjects of extensive scientific research. It has been suggested that CD83 is not only a ubiquitous co-stimulatory molecule but also a crucial player in monitoring and resolving inflammatory reactions. Although CD83 has been involved in immunological responses, its functions in autoimmune diseases and effects on pathogen immune evasion remain unclear. Herein, we outline current immunological findings and the proposed function of CD83 in inflammatory disorders.
Collapse
Affiliation(s)
- Bushra Riaz
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - S. M. Shamsul Islam
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Hye Myung Ryu
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Seonghyang Sohn
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Correspondence:
| |
Collapse
|
14
|
Yang M, Tian S, Lin Z, Fu Z, Li C. Costimulatory and coinhibitory molecules of B7-CD28 family in cardiovascular atherosclerosis: A review. Medicine (Baltimore) 2022; 101:e31667. [PMID: 36397436 PMCID: PMC9666218 DOI: 10.1097/md.0000000000031667] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Accumulating evidence supports the active involvement of vascular inflammation in atherosclerosis pathogenesis. Vascular inflammatory events within atherosclerotic plaques are predominated by innate antigen-presenting cells (APCs), including dendritic cells, macrophages, and adaptive immune cells such as T lymphocytes. The interaction between APCs and T cells is essential for the initiation and progression of vascular inflammation during atherosclerosis formation. B7-CD28 family members that provide either costimulatory or coinhibitory signals to T cells are important mediators of the cross-talk between APCs and T cells. The balance of different functional members of the B7-CD28 family shapes T cell responses during inflammation. Recent studies from both mouse and preclinical models have shown that targeting costimulatory molecules on APCs and T cells may be effective in treating vascular inflammatory diseases, especially atherosclerosis. In this review, we summarize recent advances in understanding how APC and T cells are involved in the pathogenesis of atherosclerosis by focusing on B7-CD28 family members and provide insight into the immunotherapeutic potential of targeting B7-CD28 family members in atherosclerosis.
Collapse
Affiliation(s)
- Mao Yang
- Department of Cardiology, Electrophysiological Center of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Simeng Tian
- Basic Medicine College, Harbin Medical University, Harbin, China
| | - Zhoujun Lin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Zhenkun Fu
- Basic Medicine College, Harbin Medical University, Harbin, China
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
- Department of Immunology, Wu Lien-Teh Institute, Heilongjiang Provincial Key Laboratory for Infection and Immunity, Harbin Medical University, Heilongjiang Academy of Medical Science, Harbin, China
- * Correspondence: Zhenkun Fu, Basic Medicine College, Harbin Medical University, Harbin, China (e-mail. ); Chenggang Li, State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China (e-mail. )
| | - Chenggang Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
- * Correspondence: Zhenkun Fu, Basic Medicine College, Harbin Medical University, Harbin, China (e-mail. ); Chenggang Li, State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China (e-mail. )
| |
Collapse
|
15
|
Hu Y, Hudson WH, Kissick HT, Medina CB, Baptista AP, Ma C, Liao W, Germain RN, Turley SJ, Zhang N, Ahmed R. TGF-β regulates the stem-like state of PD-1+ TCF-1+ virus-specific CD8 T cells during chronic infection. J Exp Med 2022; 219:e20211574. [PMID: 35980386 PMCID: PMC9393409 DOI: 10.1084/jem.20211574] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 06/01/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Recent studies have defined a novel population of PD-1+ TCF-1+ stem-like CD8 T cells in chronic infections and cancer. These quiescent cells reside in lymphoid tissues, are critical for maintaining the CD8 T cell response under conditions of persistent antigen, and provide the proliferative burst after PD-1 blockade. Here we examined the role of TGF-β in regulating the differentiation of virus-specific CD8 T cells during chronic LCMV infection of mice. We found that TGF-β signaling was not essential for the generation of the stem-like CD8 T cells but was critical for maintaining the stem-like state and quiescence of these cells. TGF-β regulated the unique transcriptional program of the stem-like subset, including upregulation of inhibitory receptors specifically expressed on these cells. TGF-β also promoted the terminal differentiation of exhausted CD8 T cells by suppressing the effector-associated program. Together, the absence of TGF-β signaling resulted in significantly increased accumulation of effector-like CD8 T cells. These findings have implications for immunotherapies in general and especially for T cell therapy against chronic infections and cancer.
Collapse
Affiliation(s)
- Yinghong Hu
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | - William H. Hudson
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | - Haydn T. Kissick
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
- Winship Cancer Institute of Emory University, Atlanta, GA
- Department of Urology, Emory University School of Medicine, Atlanta, GA
| | - Christopher B. Medina
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | - Antonio P. Baptista
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGhent Center for Inflammation Research, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Chaoyu Ma
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Wei Liao
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ronald N. Germain
- Lymphocyte Biology Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | | | - Nu Zhang
- Department of Microbiology, Immunology and Molecular Genetics, Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
16
|
Royzman D, Peckert-Maier K, Stich L, König C, Wild AB, Tauchi M, Ostalecki C, Kiesewetter F, Seyferth S, Lee G, Eming SA, Fuchs M, Kunz M, Stürmer EK, Peters EMJ, Berking C, Zinser E, Steinkasserer A. Soluble CD83 improves and accelerates wound healing by the induction of pro-resolving macrophages. Front Immunol 2022; 13:1012647. [PMID: 36248909 PMCID: PMC9564224 DOI: 10.3389/fimmu.2022.1012647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022] Open
Abstract
To facilitate the recovery process of chronic and hard-to-heal wounds novel pro-resolving treatment options are urgently needed. We investigated the pro-regenerative properties of soluble CD83 (sCD83) on cutaneous wound healing, where sCD83 accelerated wound healing not only after systemic but also after topical application, which is of high therapeutic interest. Cytokine profile analyses revealed an initial upregulation of inflammatory mediators such as TNFα and IL-1β, followed by a switch towards pro-resolving factors, including YM-1 and IL-10, both expressed by tissue repair macrophages. These cells are known to mediate resolution of inflammation and stimulate wound healing processes by secretion of growth factors such as epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF), which promote vascularization as well as fibroblast and keratinocyte differentiation. In conclusion, we have found strong wound healing capacities of sCD83 beyond the previously described role in transplantation and autoimmunity. This makes sCD83 a promising candidate for the treatment of chronic- and hard-to-heal wounds.
Collapse
Affiliation(s)
- Dmytro Royzman
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- *Correspondence: Dmytro Royzman, ; Alexander Steinkasserer,
| | - Katrin Peckert-Maier
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christina König
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andreas B. Wild
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Miyuki Tauchi
- Department of Internal Medicine 2, University Hospital Erlangen, FAU, Erlangen, Germany
| | - Christian Ostalecki
- Department of Dermatology, University Hospital Erlangen, FAU, Erlangen, Germany
| | | | - Stefan Seyferth
- Division of Pharmaceutics, Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Geoffrey Lee
- Division of Pharmaceutics, Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sabine A. Eming
- Department of Dermatology, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne Excellence Cluster Cluster of Excellence for Aging Research (CECAD), University of Cologne, Cologne, Germany
| | - Maximilian Fuchs
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Meik Kunz
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
- Department of Medical Informatics, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Ewa K. Stürmer
- Department for Vascular Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva M. J. Peters
- Psychoneuroimmunology Laboratory, Klinik für Psychosomatik und Psychotherapie, Justus-Liebig Universität Gießen, Gießen, Germany
| | - Carola Berking
- Department of Dermatology, University Hospital Erlangen, FAU, Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- *Correspondence: Dmytro Royzman, ; Alexander Steinkasserer,
| |
Collapse
|
17
|
Royzman D, Andreev D, Stich L, Peckert-Maier K, Wild AB, Zinser E, Mühl-Zürbes P, Jones E, Adam S, Frey S, Fuchs M, Kunz M, Bäuerle T, Nagel L, Schett G, Bozec A, Steinkasserer A. The soluble CD83 protein prevents bone destruction by inhibiting the formation of osteoclasts and inducing resolution of inflammation in arthritis. Front Immunol 2022; 13:936995. [PMID: 36003376 PMCID: PMC9393726 DOI: 10.3389/fimmu.2022.936995] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/08/2022] [Indexed: 12/29/2022] Open
Abstract
Here we show that soluble CD83 induces the resolution of inflammation in an antigen-induced arthritis (AIA) model. Joint swelling and the arthritis-related expression levels of IL-1β, IL-6, RANKL, MMP9, and OC-Stamp were strongly reduced, while Foxp3 was induced. In addition, we observed a significant inhibition of TRAP+ osteoclast formation, correlating with the reduced arthritic disease score. In contrast, cell-specific deletion of CD83 in human and murine precursor cells resulted in an enhanced formation of mature osteoclasts. RNA sequencing analyses, comparing sCD83- with mock treated cells, revealed a strong downregulation of osteoclastogenic factors, such as Oc-Stamp, Mmp9 and Nfatc1, Ctsk, and Trap. Concomitantly, transcripts typical for pro-resolving macrophages, e.g., Mrc1/2, Marco, Klf4, and Mertk, were upregulated. Interestingly, members of the metallothionein (MT) family, which have been associated with a reduced arthritic disease severity, were also highly induced by sCD83 in samples derived from RA patients. Finally, we elucidated the sCD83-induced signaling cascade downstream to its binding to the Toll-like receptor 4/(TLR4/MD2) receptor complex using CRISPR/Cas9-induced knockdowns of TLR4/MyD88/TRIF and MTs, revealing that sCD83 acts via the TRIF-signaling cascade. In conclusion, sCD83 represents a promising therapeutic approach to induce the resolution of inflammation and to prevent bone erosion in autoimmune arthritis.
Collapse
Affiliation(s)
- Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Dmytro Royzman, ; Alexander Steinkasserer,
| | - Darja Andreev
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Katrin Peckert-Maier
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas B. Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Evan Jones
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Adam
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Silke Frey
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Maximilian Fuchs
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Meik Kunz
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
- Department of Medical Informatics, Friedrich-Alexander University (FAU) of Erlangen-Nürnberg, Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Lisa Nagel
- Institute of Radiology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- *Correspondence: Dmytro Royzman, ; Alexander Steinkasserer,
| |
Collapse
|
18
|
Evaluation of Soluble CD90: Potential for Diagnostic Significance in Endometriosis Patients. DISEASE MARKERS 2022; 2022:9345858. [PMID: 35769819 PMCID: PMC9236764 DOI: 10.1155/2022/9345858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/04/2022] [Indexed: 11/25/2022]
Abstract
Background Endometriosis is a chronic and debilitating gynecologic disorder, driven by endocrine and immune dysfunctions, which lead to poor endometrial differentiation and attenuated fertility. Escape from immune surveillance and involvement of inflammatory mechanisms appear to be factors in disease progression. Current diagnostic guidelines for endometriosis still lack an efficient biomarker. Here, we report a study on two previously unexplored factors as potential biomarkers for endometriosis. Methods A case-control study was performed to evaluate the diagnostic potential of serum CD90 and CD83 levels in endometriosis patients (cases validated by surgical and histological examination) compared to healthy controls. Serum was collected from age-matched females and analyzed by ELISA. Results Comparison of endometriosis patients to the control group showed significantly elevated levels of serum CD90 (1160 ± 856 pg/mL vs. 334 ± 228 pg/mL; ∗∗∗∗p < 0.0001). A threshold value of 479.4 pg/mL was defined based on the control results, and the diagnostic efficiency of the test was estimated. The obtained sensitivity (70.4%), specificity (92.9%), positive predictive value (90.5%), and negative predictive value (76.5%) rated the test as one with promising diagnostic potential. In contrast, the analysis of serum CD83 levels showed comparable values in both groups, suggesting no association with patient status. Conclusion Elevated soluble CD90 in human serum is associated with endometriosis, which suggests its putative clinical significance as a biomarker in screening and/or diagnosis of the disease.
Collapse
|
19
|
Peckert-Maier K, Schönberg A, Wild AB, Royzman D, Braun G, Stich L, Hadrian K, Tripal P, Cursiefen C, Steinkasserer A, Zinser E, Bock F. Pre-incubation of corneal donor tissue with sCD83 improves graft survival via the induction of alternatively activated macrophages and tolerogenic dendritic cells. Am J Transplant 2022; 22:438-454. [PMID: 34467638 DOI: 10.1111/ajt.16824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/02/2021] [Accepted: 08/22/2021] [Indexed: 01/25/2023]
Abstract
Immune responses reflect a complex interplay of cellular and extracellular components which define the microenvironment of a tissue. Therefore, factors that locally influence the microenvironment and re-establish tolerance might be beneficial to mitigate immune-mediated reactions, including the rejection of a transplant. In this study, we demonstrate that pre-incubation of donor tissue with the immune modulator soluble CD83 (sCD83) significantly improves graft survival using a high-risk corneal transplantation model. The induction of tolerogenic mechanisms in graft recipients was achieved by a significant upregulation of Tgfb, Foxp3, Il27, and Il10 in the transplant and an increase of regulatory dendritic cells (DCs), macrophages (Mφ), and T cells (Tregs) in eye-draining lymph nodes. The presence of sCD83 during in vitro DC and Mφ generation directed these cells toward a tolerogenic phenotype leading to reduced proliferation-stimulating activity in MLRs. Mechanistically, sCD83 induced a tolerogenic Mφ and DC phenotype, which favors Treg induction and significantly increased transplant survival after adoptive cell transfer. Conclusively, pre-incubation of corneal grafts with sCD83 significantly prolongs graft survival by modulating recipient Mφ and DCs toward tolerance and thereby establishing a tolerogenic microenvironment. This functional strategy of donor graft pre-treatment paves the way for new therapeutic options in the field of transplantation.
Collapse
Affiliation(s)
- Katrin Peckert-Maier
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alfrun Schönberg
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Andreas B Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gabriele Braun
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Karina Hadrian
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Tripal
- Optical Imaging Centre, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Claus Cursiefen
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Felix Bock
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
20
|
Probiotic Molecules That Inhibit Inflammatory Diseases. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Consumption of probiotics for health purposes has increased vastly in the past few decades, and yet the scientific evidence to support health benefits from probiotics is only beginning to emerge. As more probiotics are studied, we are beginning to understand the mechanisms of action by which they benefit human health, as well as to identify the bacterial molecules responsible for these benefits. A new era of therapeutics is on the horizon in which purified molecules from probiotics will be used to prevent and treat diseases. In this review, we summarize the active molecules from probiotic bacteria that have been shown to affect innate and adaptive immunity and have health benefits in experimental settings. We focus particularly on the cellular and molecular mechanisms of the probiotic Bacillus subtilis and its active molecule, exopolysaccharide (ESPBs).
Collapse
|
21
|
Peckert-Maier K, Royzman D, Langguth P, Marosan A, Strack A, Sadeghi Shermeh A, Steinkasserer A, Zinser E, Wild AB. Tilting the Balance: Therapeutic Prospects of CD83 as a Checkpoint Molecule Controlling Resolution of Inflammation. Int J Mol Sci 2022; 23:732. [PMID: 35054916 PMCID: PMC8775349 DOI: 10.3390/ijms23020732] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammatory diseases and transplant rejection represent major challenges for modern health care. Thus, identification of immune checkpoints that contribute to resolution of inflammation is key to developing novel therapeutic agents for those conditions. In recent years, the CD83 (cluster of differentiation 83) protein has emerged as an interesting potential candidate for such a "pro-resolution" therapy. This molecule occurs in a membrane-bound and a soluble isoform (mCD83 and sCD83, respectively), both of which are involved in resolution of inflammation. Originally described as a maturation marker on dendritic cells (DCs), mCD83 is also expressed by activated B and T cells as well as regulatory T cells (Tregs) and controls turnover of MHC II molecules in the thymus, and thereby positive selection of CD4+ T cells. Additionally, it serves to confine overshooting (auto-)immune responses. Consequently, animals with a conditional deletion of CD83 in DCs or regulatory T cells suffer from impaired resolution of inflammation. Pro-resolving effects of sCD83 became evident in pre-clinical autoimmune and transplantation models, where application of sCD83 reduced disease symptoms and enhanced allograft survival, respectively. Here, we summarize recent advances regarding CD83-mediated resolution of inflammatory responses, its binding partners as well as induced signaling pathways, and emphasize its therapeutic potential for future clinical trials.
Collapse
Affiliation(s)
- Katrin Peckert-Maier
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander Universität—Erlangen-Nürnberg, 91052 Erlangen, Germany; (D.R.); (P.L.); (A.M.); (A.S.); (A.S.S.); (A.S.); (E.Z.)
| | | | | | | | | | | | | | | | - Andreas B. Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander Universität—Erlangen-Nürnberg, 91052 Erlangen, Germany; (D.R.); (P.L.); (A.M.); (A.S.); (A.S.S.); (A.S.); (E.Z.)
| |
Collapse
|
22
|
Talker SC, Barut GT, Lischer HE, Rufener R, von Münchow L, Bruggmann R, Summerfield A. Monocyte biology conserved across species: Functional insights from cattle. Front Immunol 2022; 13:889175. [PMID: 35967310 PMCID: PMC9373011 DOI: 10.3389/fimmu.2022.889175] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 06/30/2022] [Indexed: 12/24/2022] Open
Abstract
Similar to human monocytes, bovine monocytes can be split into CD14highCD16- classical, CD14highCD16high intermediate and CD14-/dimCD16high nonclassical monocytes (cM, intM, and ncM, respectively). Here, we present an in-depth analysis of their steady-state bulk- and single-cell transcriptomes, highlighting both pronounced functional specializations and transcriptomic relatedness. Bulk gene transcription indicates pro-inflammatory and antibacterial roles of cM, while ncM and intM appear to be specialized in regulatory/anti-inflammatory functions and tissue repair, as well as antiviral responses and T-cell immunomodulation. Notably, intM stood out by high expression of several genes associated with antigen presentation. Anti-inflammatory and antiviral functions of ncM are further supported by dominant oxidative phosphorylation and selective strong responses to TLR7/8 ligands, respectively. Moreover, single-cell RNA-seq revealed previously unappreciated heterogeneity within cM and proposes intM as a transient differentiation intermediate between cM and ncM.
Collapse
Affiliation(s)
- Stephanie C. Talker
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- *Correspondence: Stephanie C. Talker,
| | - G. Tuba Barut
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Heidi E.L. Lischer
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Reto Rufener
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Rémy Bruggmann
- Interfaculty Bioinformatics Unit and Swiss Institute of Bioinformatics, University of Bern, Bern, Switzerland
| | - Artur Summerfield
- Institute of Virology and Immunology, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Biele E, Schober SJ, Prexler C, Thiede M, von Heyking K, Gassmann H, Eck J, Xue B, Burdach S, Thiel U. Monocyte Maturation Mediators Upregulate CD83, ICAM-1 and MHC Class 1 Expression on Ewing's Sarcoma, Enhancing T Cell Cytotoxicity. Cells 2021; 10:3070. [PMID: 34831294 PMCID: PMC8624504 DOI: 10.3390/cells10113070] [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: 09/30/2021] [Revised: 10/22/2021] [Accepted: 11/05/2021] [Indexed: 11/29/2022] Open
Abstract
Ewing's sarcoma (EwS) is a pediatric solid tumor entity with low somatic mutational burden and a low rate of tumor-infiltrating T cells, indicating a low extent of immunogenicity. In EwS, immunogenicity may furthermore be significantly diminished by a predominantly M2 macrophage driven pro-tumorigenic tumor microenvironment. In the past, we demonstrated that CHM1319-specific TCR-transgenic T cells are able to control EwS growth in a preclinical mouse model as well as in a patient with metastatic disease. However, new adjuvant techniques to induce long lasting and curative CHM1319-specific TCR-transgenic T cell-mediated anti-tumor responses are needed. In this work, we sought to identify a technique to improve the cytotoxic effect of CHM1319-specific TCR-transgenic T cell by altering the immunogenic cell surface marker expression on EwS cell lines using different cytokines. We demonstrate that TNF, IL-6, IL-1β and PGE2 cause pro-immunogenic CD83, MHC class I and II as well as ICAM-1 upregulation in EwS cell lines. This observation was associated with significantly improved recognition and killing of the tumor cells by EwS-specific CHM1319/HLA-A*02:01-restricted TCR-transgenic T cells. Conclusively, we demonstrate that the induction of an inflammatory signature renders EwS more susceptible to adoptive T cell therapy. TNF, which is upregulated during inflammatory processes, is of particular translational interest as its secretion may be induced in the patients e.g., by irradiation and hyperthermia in the clinical setting. In future clinical protocols, this finding may be important to identify appropriate conditioning regimens as well as point of time for adoptive T cell-based immunotherapy in EwS patients.
Collapse
Affiliation(s)
- Emilie Biele
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| | - Sebastian J. Schober
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| | - Carolin Prexler
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| | - Melanie Thiede
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| | - Kristina von Heyking
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| | - Hendrik Gassmann
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| | - Jennifer Eck
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| | - Busheng Xue
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| | - Stefan Burdach
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
- German Cancer Consortium (DKTK), German Research Center (DKFZ), Partner Site Munich, 80336 Munich, Germany
| | - Uwe Thiel
- Department of Pediatrics, Children’s Cancer Research Center, Kinderklinik München Schwabing, School of Medicine, Technical University of Munich, 80804 Munich, Germany; (S.J.S.); (C.P.); (M.T.); (K.v.H.); (H.G.); (J.E.); (B.X.); (S.B.)
| |
Collapse
|
24
|
Zhao LR, Xiao J, Shang Q, Li T, Liu XS, Guan FL. Application of CD83 and HSF5 to Identify Antemortem and Postmortem Skin Burns. FA YI XUE ZA ZHI 2021; 37:627-631. [PMID: 35187913 DOI: 10.12116/j.issn.1004-5619.2020.400401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
OBJECTIVES To explore the forensic application value of cluster of differentiation 83 (CD83) and heat shock transcription factor 5(HSF5) in identifying antemortem and postmortem skin burns. METHODS Through reverse transcription real-time quantitative polymerase chain reaction (RT-qPCR), CD83 and HSF5 mRNA levels in the skin tissues of antemortem and postmortem burned mice and human samples were detected quantitatively. RESULTS Compared with the control group and the postmortem burned group, the mRNA levels of CD83 and HSF5 in antemortem burned mice were higher. The high mRNA expressions of CD83 could be detected 96 h after death, and the mRNA expressions of HSF5 could be observed 72 h after death. Compared with undamaged skin, increased CD83 and HSF5 mRNA levels were detected in 11 out of 15 cases(P<0.05). CONCLUSIONS CD83 and HSF5 can be used in forensic practice as indicators for vital reaction in antemortem burn identification.
Collapse
Affiliation(s)
- Long-Rui Zhao
- School of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Jing Xiao
- School of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Qing Shang
- School of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Tao Li
- School of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Xin-She Liu
- School of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Fang-Lin Guan
- School of Forensic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| |
Collapse
|
25
|
Puck A, Künig S, Modak M, May L, Fritz P, Battin C, Radakovics K, Steinberger P, Reipert BM, Crowe BA, Stöckl J. The soluble cytoplasmic tail of CD45 regulates T-cell activation via TLR4 signaling. Eur J Immunol 2021; 51:3176-3185. [PMID: 34626426 DOI: 10.1002/eji.202149227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/03/2021] [Accepted: 10/06/2021] [Indexed: 11/10/2022]
Abstract
The soluble cytoplasmic tail of CD45 (ct-CD45) is a cleavage fragment of CD45, that is generated during the activation of human phagocytes. Upon release to the extracellular space, ct-CD45 binds to human T cells and inhibits their activation in vitro. Here, we studied the potential role of TLR4 as a receptor for ct-CD45. Treatment of Jurkat TLR4/CD14 reporter cells with ct-CD45 induced the upregulation of the reporter gene NFκB-eGFP and could be blocked by inhibitors of TLR4 signaling. Conversely, ct-CD45 did not promote the NFκB-controlled eGFP induction in reporter cells expressing TLR1, TLR2, and TLR6 transgenes and did not lead to the activation of the transcription factors NFκB, AP-1, and NFAT in a Jurkat reporter cell line expressing endogenous TLR5. Moreover, ct-CD45 binds to recombinant TLR4 in an in vitro assay and this association was reduced in the presence of oxidized 1-palmitoyl-2-arachidonyl-sn-glycero-3-phosphorylcholine. Blockade of TLR4 with mAb HTA125 partially reversed the ct-CD45-mediated inhibition of T-cell proliferation. Interestingly, targeting of TLR4 with mAb W7C11 also suppressed T-cell proliferation. In summary, the results of this study demonstrate that ct-CD45 acts via a noncanonical TLR4 activation pathway on T cells, which modulates TCR signaling.
Collapse
Affiliation(s)
- Alexander Puck
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sarojinidevi Künig
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Madhura Modak
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Lara May
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Pia Fritz
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Claire Battin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Katharina Radakovics
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Birgit M Reipert
- Department of Immunology, Drug Discovery Austria, Baxalta Innovations GmbH, Vienna, Austria
| | - Brian A Crowe
- Department of Immunology, Drug Discovery Austria, Baxalta Innovations GmbH, Vienna, Austria
| | - Johannes Stöckl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
26
|
Mo L, Luo X, Yang G, Liu J, Yang L, Liu Z, Wang S, Liu D, Liu Z, Yang P. Epithelial cell-derived CD83 restores immune tolerance in the airway mucosa by inducing regulatory T-cell differentiation. Immunology 2021; 163:310-322. [PMID: 33539546 PMCID: PMC8207377 DOI: 10.1111/imm.13317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/06/2021] [Accepted: 01/08/2021] [Indexed: 12/11/2022] Open
Abstract
The mechanism of generation of regulatory T cells (Treg) remains incompletely understood. Recent studies show that CD83 has immune regulatory functions. This study aims to investigate the role of epithelial cell-derived CD83 in the restoration of immune tolerance in the airway mucosa by inducing the Treg differentiation. In this study, CD83 and ovalbumin (OVA)-carrying exosomes were generated from airway epithelial cells. An airway allergy mouse model was developed to test the role of CD83/OVA-carrying exosomes in the suppression of airway allergy by inducing Treg generation. We observed that mouse airway epithelial cells expressed CD83 that could be up-regulated by CD40 ligand. The CD83 deficiency in epithelial cells retarded the Treg generation in the airway mucosa. CD83 up-regulated transforming growth factor-β-inducible early gene 1 expression in CD4+ T cells to promote Foxp3 expression. Exposure of primed CD4+ T cells to CD83/OVA-carrying exosomes promoted antigen-specific Treg generation. Administration of CD83/OVA-carrying exosomes inhibited experimental airway allergic response. In summary, airway epithelial cells express CD83 that is required in the Treg differentiation in the airway mucosa. Administration of CD83/OVA-carrying exosomes can inhibit airway allergy that has the translation potential in the treatment of airway allergic disorders.
Collapse
Affiliation(s)
- Li‐Hua Mo
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
| | - Xiang‐Qian Luo
- Department of Pediatric OtolaryngologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Gui Yang
- Department of OtolaryngologyLonggang Central HospitalShenzhenChina
| | - Jiang‐Qi Liu
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Li‐Teng Yang
- Department of Respirology & AllergyThird Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Zhi‐Qiang Liu
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Shuai Wang
- Longgang ENT Hospital & Shenzhen ENT InstituteShenzhenChina
| | - Da‐Bo Liu
- Department of Pediatric OtolaryngologyShenzhen HospitalSouthern Medical UniversityShenzhenChina
| | - Zhi‐Gang Liu
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
| | - Ping‐Chang Yang
- Research Center of Allergy & ImmunologyShenzhen University School of MedicineShenzhenChina
- Guangdong Provincial Key Laboratory of Regional Immunity and DiseasesShenzhenChina
| |
Collapse
|
27
|
Zhang R, Song YN, Duo X, Guo Z, Sun Y, Zhang Z, Lu Y, Miao B, Yang PC, Nie G. Retinoblastoma cell-derived Twist protein promotes regulatory T cell development. Cancer Immunol Immunother 2021; 70:1037-1048. [PMID: 33108472 PMCID: PMC10992014 DOI: 10.1007/s00262-020-02744-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/12/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND The development of tumor tissue-infiltrating regulatory T cell (Treg) is incompletely understood. This study investigates the role of retinoblastoma cell (Rbc)-derived Twist‑related protein 1 (Twist) in the Treg development. METHODS The surgically removed Rb tissues were collected. Rbcs were cultured with CD4+ T cells to assess the role of Rbc-derived Twist in the Treg generation. RESULTS We found that more than 90% Rbcs expressed Twist. Foxp3+ Tregs were detected in the Rb tissues that were positively correlated with the Twist expression in Rbcs, negatively associated with Rb patient survival and sight survival. Treating Rbcs with hypoxia promoted the Twist expression that could be detected in the cytoplasm, nuclei and on the cell surface. Twist activated CD4+ T cells by binding the TLR4/myeloid differentiation factor 2 complex and promoted the transforming growth factor-β-inducible early gene 1 product and Foxp3 expression. These Rbc-induced Foxp3+ Tregs showed immune-suppressive function on CD8+ T cell proliferation. CONCLUSIONS Rbcs express Twist, that induces IL-4+ Foxp3+ Tregs; the latter can inhibit CD8+ cytotoxic T cell activities. Therefore, Twist may play an important role in the pathogenesis of Rb.
Collapse
Affiliation(s)
- Ruishi Zhang
- Department of Ophthalmology, Shenzhen Secondary Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yan-Nan Song
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Room A7-509, Lihu Campus, 1066 Xueyuan Blvd, Shenzhen, 518055, China
| | - Xiaoyan Duo
- Department of Ophthalmology, Shenzhen Secondary Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhihong Guo
- Shenzhen Luohu Medical Group, Shenzhen, China
| | - Yanhua Sun
- Department of Pathology, Shenzhen Secondary Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhixiong Zhang
- Department of Pathology, Shenzhen Secondary Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yongtian Lu
- Department of Otolaryngology, Head and Neck Surgery, Shenzhen Secondary Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Beiping Miao
- Department of Otolaryngology, Head and Neck Surgery, Shenzhen Secondary Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| | - Ping-Chang Yang
- Research Center of Allergy and Immunology, Shenzhen University School of Medicine, Room A7-509, Lihu Campus, 1066 Xueyuan Blvd, Shenzhen, 518055, China.
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China.
| | - Guohui Nie
- Department of Otolaryngology, Head and Neck Surgery, Shenzhen Secondary Hospital and First Affiliated Hospital of Shenzhen University, Shenzhen, China.
| |
Collapse
|
28
|
Suttapitugsakul S, Tong M, Sun F, Wu R. Enhancing Comprehensive Analysis of Secreted Glycoproteins from Cultured Cells without Serum Starvation. Anal Chem 2021; 93:2694-2705. [PMID: 33397101 PMCID: PMC8034805 DOI: 10.1021/acs.analchem.0c05126] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glycoproteins secreted by cells play essential roles in the regulation of extracellular activities. Secreted glycoproteins are often reflective of cellular status, and thus glycoproteins from easily accessible bodily fluids can serve as excellent biomarkers for disease detection. Cultured cells have been extensively employed as models in the research fields of biology and biomedicine, and global analysis of glycoproteins secreted from these cells provides insights into cellular activities and glycoprotein functions. However, comprehensive identification and quantification of secreted glycoproteins is a daunting task because of their low abundances compared with the high-abundance serum proteins required for cell growth and proliferation. Several studies employed serum-free media to analyze secreted proteins, but it has been shown that serum starvation, even for a short period of time, can alter protein secretion. To overcome these issues, we developed a method to globally characterize secreted glycoproteins and their N-glycosylation sites from cultured cells by combining selective enrichment of secreted glycoproteins with a boosting approach. The results demonstrated the importance of the boosting sample selection and the boosting-to-sample ratio for improving the coverage of secreted glycoproteins. The method was applied to globally quantify secreted glycoproteins from THP-1 monocytes and macrophages in response to lipopolysaccharides (LPS) and from Hep G2 cells treated with TGF-β without serum starvation. We found differentially secreted glycoproteins in these model systems that showed the cellular response to the immune activation or the epithelial-to-mesenchymal transition. Benefiting from the selective enrichment and the signal enhancement of low-abundance secreted glycoproteins, this method can be extensively applied to study secreted glycoproteins without serum starvation, which will provide a better understanding of protein secretion and cellular activity.
Collapse
Affiliation(s)
- Suttipong Suttapitugsakul
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Ming Tong
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Fangxu Sun
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Ronghu Wu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| |
Collapse
|
29
|
Lin W, Zhou S, Feng M, Yu Y, Su Q, Li X. Soluble CD83 Regulates Dendritic Cell-T Cell Immunological Synapse Formation by Disrupting Rab1a-Mediated F-Actin Rearrangement. Front Cell Dev Biol 2021; 8:605713. [PMID: 33585445 PMCID: PMC7874230 DOI: 10.3389/fcell.2020.605713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/11/2020] [Indexed: 12/23/2022] Open
Abstract
Dendritic cell-T cell (DC-T) contacts play an important role in T cell activation, clone generation, and development. Regulating the cytoskeletal protein rearrangement of DCs can modulate DC-T contact and affect T cell activation. However, inhibitory factors on cytoskeletal regulation in DCs remain poorly known. We showed that a soluble form of CD83 (sCD83) inhibited T cell activation by decreasing DC-T contact and synapse formation between DC and T cells. This negative effect of sCD83 on DCs was mediated by disruption of F-actin rearrangements, leading to alter expression and localization of major histocompatibility complex class II (MHC-II) and immunological synapse formation between DC and T cells. Furthermore, sCD83 was found to decrease GTP-binding activity of Rab1a, which further decreased colocalization and expression of LRRK2 and F-actin rearrangements in DCs, leading to the loss of MHC-II at DC-T synapses and reduced DC-T synapse formation. Further, sCD83-treated DCs alleviated symptoms of experimental autoimmune uveitis in mice and decreased the number of T cells in the eyes and lymph nodes of these animals. Our findings demonstrate a novel signaling pathway of sCD83 on regulating DC-T contact, which may be harnessed to develop new immunosuppressive therapeutics for autoimmune disease.
Collapse
Affiliation(s)
- Wei Lin
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, China
| | - Shuping Zhou
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, China
| | - Meng Feng
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, China
| | - Yong Yu
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, China
| | - Qinghong Su
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, China
| | - Xiaofan Li
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, China
| |
Collapse
|
30
|
Nakatomi K, Ueno H, Ishikawa Y, Salim RC, Mori Y, Kanemoto I, Tancharoen S, Kikuchi K, Miura N, Omori T, Okuda-Ashitaka E, Matsumura K, Imaizumi H, Motomiya Y, Maruyama I, Kawahara KI. TLR4/MD-2 is a receptor for extracellular nucleophosmin 1. Biomed Rep 2020; 14:21. [PMID: 33335727 PMCID: PMC7739869 DOI: 10.3892/br.2020.1397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022] Open
Abstract
Nucleophosmin 1 (NPM1) primarily localizes to the nucleus and is passively released into the extracellular milieu by necrotic or damaged cells, or is secreted by monocytes and macrophages. Extracellular NPM1 acts as a potent inflammatory stimulator by promoting cytokine production [e.g., tumor necrosis factor-α (TNF-α)], which suggests that NPM1 acts as a damage-associated molecular pattern. However, the receptor of NPM1 is unknown. Evidence indicates that DAMPs, which include high mobility group box 1 and histones, may bind Toll-like receptors (TLRs). In the present study, it was shown that NPM1 signaling was mediated via the TLR4 pathway, which suggests that TLR4 is an NPM1 receptor. TLR4 binds myeloid differentiation protein-2 (MD-2), which is essential for intracellular signaling. Furthermore, the TLR4 antagonist, LPS-Rhodobacter sphaeroides (an MD-2 antagonist) and TAK-242 (a TLR4 signaling inhibitor) significantly inhibited NPM1-induced TNF-α production by differentiated THP-1 cells as well as reducing ERK1/2 activation. Far-western blot analysis revealed that NPM1 directly bound MD-2. Thus, the results of the present study provide compelling evidence that TLR4 binds NPM1, and it is hypothesized that inhibiting NPM1 activity may serve as a novel strategy for treating TLR4-related diseases.
Collapse
Affiliation(s)
- Kota Nakatomi
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Hikari Ueno
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Yuto Ishikawa
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Ronny Christiadi Salim
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Yuki Mori
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Issey Kanemoto
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Salunya Tancharoen
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan.,Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand
| | - Kiyoshi Kikuchi
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok 10400, Thailand.,Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan.,Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8520, Japan
| | - Naoki Miura
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-8580, Japan
| | - Taketo Omori
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Emiko Okuda-Ashitaka
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Kiyoshi Matsumura
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan
| | - Hitoshi Imaizumi
- Department of Anesthesiology and Intensive Care Medicine, Tokyo Medical University, School of Medicine, Tokyo 160-0023, Japan
| | | | - Ikuro Maruyama
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan.,Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8520, Japan
| | - Ko-Ichi Kawahara
- Department of Biomedical Engineering, Osaka Institute of Technology, Osaka 535-8585, Japan.,Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima 890-8520, Japan
| |
Collapse
|
31
|
Džopalić T, Kostić M, Kostić M, Marjanović G, Guzina J, Jurišić V, Božić Nedeljković B. Effects of galectin-1 on immunomodulatory properties of human monocyte-derived dendritic cells. Growth Factors 2020; 38:235-246. [PMID: 34223785 DOI: 10.1080/08977194.2021.1947267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Our study aimed to evaluate the effects of Gal-1 in dose depending manner on maturation and immunomodulatory properties of monocyte-derived (Mo) DCs in-vitro. The effects were analyzed by monitoring their phenotypic characteristics, cytokine profile, and the ability to direct the immune response in the co-culture with allogeneic CD4+T cells. Gal-1 reduced the expression of CD80 and CD86 molecules on MoDCs compared to untreated MoDCs. Gal-1 at concentrations of 1 and 6 μg/mL significantly reduced IL-12 production, while the concentration of 3 μg/mL led to its significant increase. Gal-1 in all concentrations induced a significant increase in the production of IL-10. Treatment of MoDCs with 3 and 6 μg/mL of Gal-1 stimulated the production of IL-2 and IFN-γ in the co-culture with CD4+T lymphocytes. This study demonstrated a dual immunomodulatory effect of Gal-1 on MoDCs in terms of immune stimulation and immune suppression, depending on the applied concentration.
Collapse
Affiliation(s)
- Tanja Džopalić
- Department of Immunology, Medical Faculty, University of Niš, Niš, Serbia
| | - Miloš Kostić
- Department of Immunology, Medical Faculty, University of Niš, Niš, Serbia
| | - Milena Kostić
- Faculty of Biology, Institute for Physiology and Biochemistry "Ivan Djaja", University of Belgrade, Belgrade, Serbia
| | - Goran Marjanović
- Department of Immunology, Medical Faculty, University of Niš, Niš, Serbia
- Clinic for Hematology and Clinical Immunology, Clinical Center Niš, Niš, Serbia
| | - Jelena Guzina
- Faculty of Biology, Institute for Physiology and Biochemistry "Ivan Djaja", University of Belgrade, Belgrade, Serbia
| | - Vladimir Jurišić
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Biljana Božić Nedeljković
- Faculty of Biology, Institute for Physiology and Biochemistry "Ivan Djaja", University of Belgrade, Belgrade, Serbia
| |
Collapse
|
32
|
Batool A, Liu H, Liu YX, Chen SR. CD83, a Novel MAPK Signaling Pathway Interactor, Determines Ovarian Cancer Cell Fate. Cancers (Basel) 2020; 12:cancers12082269. [PMID: 32823589 PMCID: PMC7465057 DOI: 10.3390/cancers12082269] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
Ovarian cancer is a leading cause of death from gynecologic malignancies worldwide. Although CD83 is widely described as a solid marker for mature dendritic cells, emerging pieces of evidence indicate the expression of membrane protein CD83 by various tumor cells, including ovarian cancer cells. However, the potential role of CD83 in ovarian cancer cell properties and development remains absolutely unknown. By using human CD83 stable overexpression and knockdown sublines of several ovarian cancer cells, we observed that CD83 advanced the growth proliferation, colony formation ability, spheroid formation, and in vivo tumorigenicity of ovarian cancer cells; surprisingly, CD83 limited their migration and invasion potentials. Positive regulation of proliferation/stemness factors (e.g., cyclin-CDKs and KIT/CD44) but negative regulation of matrix metallopeptidases (e.g., MMP1 and 7) by CD83 were revealed by the integrated analysis of transcriptome and proteome. Furthermore, immunoprecipitation-mass spectrometry (IP-MS) and co-immunoprecipitation (Co-IP) first identified the association of CD83 with MAP3K7 (also known as TAK1) and MAP3K7-binding protein TAB1 on the cell membrane. Moreover, CD83 functions through the activation of MAP3K7-MEK1/2-ERK1/2 cascades to further regulate downstream FOXO1/p21/CDK2/CCNB1 and STAT3/DKK1 signaling pathways, thus activating proliferation and spheroid formation of ovarian cancer cells, respectively. Collectively, our findings define a CD83-MAPK pathway in the regulation of proliferation and stemness in ovarian cancer cells, with potential therapeutic applications in blocking their progression.
Collapse
Affiliation(s)
- Aalia Batool
- Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China; (A.B.); (H.L.)
- Laboratory of Reproductive Neuroendocrinology, Department of Zoology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad 45320, Pakistan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Hao Liu
- Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China; (A.B.); (H.L.)
| | - Yi-Xun Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Su-Ren Chen
- Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, Beijing 100875, China; (A.B.); (H.L.)
- Correspondence:
| |
Collapse
|
33
|
Liedtke K, Alter C, Günther A, Hövelmeyer N, Klopfleisch R, Naumann R, Wunderlich FT, Buer J, Westendorf AM, Hansen W. Endogenous CD83 Expression in CD4 + Conventional T Cells Controls Inflammatory Immune Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:3217-3226. [PMID: 32341061 DOI: 10.4049/jimmunol.2000042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/10/2020] [Indexed: 12/16/2022]
Abstract
The glycoprotein CD83 is known to be expressed by different immune cells including activated CD4+Foxp3+ regulatory T cells (Tregs) and CD4+Foxp3- conventional T cells. However, the physiological function of endogenous CD83 in CD4+ T cell subsets is still unclear. In this study, we have generated a new CD83flox mouse line on BALB/c background, allowing for specific ablation of CD83 in T cells upon breeding with CD4-cre mice. Tregs from CD83flox/flox/CD4-cretg/wt mice had similar suppressive activity as Tregs from CD83flox/flox/CD4-crewt/wt wild-type littermates, suggesting that endogenous CD83 expression is dispensable for the inhibitory capacity of Tregs. However, CD83-deficient CD4+ conventional T cells showed elevated proliferation and IFN-γ secretion as well as an enhanced capacity to differentiate into Th1 cells and Th17 cells upon stimulation in vitro. T cell-specific ablation of CD83 expression resulted in aggravated contact hypersensitivity reaction accompanied by enhanced CD4+ T cell activation. Moreover, adoptive transfer of CD4+CD45RBhigh T cells from CD83flox/flox/CD4-cretg /wt mice into Rag2-deficient mice elicited more severe colitis associated with increased serum concentrations of IL-12 and elevated CD40 expression on CD11c+ dendritic cells (DCs). Strikingly, DCs from BALB/c mice cocultured with CD83-deficient CD4+ conventional T cells showed enhanced CD40 expression and IL-12 secretion compared with DCs cocultured with CD4+ conventional T cells from CD83flox/flox/CD4-crewt/wt wild-type mice. In summary, these results indicate that endogenous CD83 expression in CD4+ conventional T cells plays a crucial role in controlling CD4+ T cell responses, at least in part, by regulating the activity of CD11c+ DCs.
Collapse
Affiliation(s)
- Katarina Liedtke
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Christina Alter
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Anne Günther
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Nadine Hövelmeyer
- Institute for Medical Medicine, University Medical Center of the Johannes-Gutenberg University Mainz, 55131 Mainz, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Free University of Berlin, 14163 Berlin, Germany
| | - Ronald Naumann
- Transgenic Core Facility, Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - F Thomas Wunderlich
- Max Planck Institute for Metabolism Research, Center for Endocrinology, Diabetes and Preventive Medicine, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany; and
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 45147 Essen, Germany;
| |
Collapse
|
34
|
Song W, Li H, Jia B, Wang Z, Liu Q, Yang G, Li X. Soluble CD83 suppresses experimental food allergy via regulating aberrant T helper 2 responses. Immunol Res 2020; 68:141-151. [PMID: 32529460 DOI: 10.1007/s12026-020-09133-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Aberrant T helper-2 (Th2) responses play a critical role in the pathogenesis of allergic diseases. The underlying mechanism is to be further investigated. It is reported that soluble CD83 (sCD83) has immune-regulatory effects. This study aims to investigate the role of sCD83 in the regulation of Th2 polarization. Blood samples were collected from pediatric patients with food allergy (FA). The Th2 response was analyzed by pertinent immunological approaches. An FA murine model was developed to test the role of sCD83 in the regulation of FA response. We found that the serum sCD83 levels were lower in FA patients. A negative correlation was detected between serum sCD83 levels and serum Th2 cytokine levels. The presence of sCD83 suppressed Th2 cell differentiation and antigen-specific Th2 cell activation. sCD83 upregulated the T-bet expression and suppressed the GATA3 expression in CD4+ T cells. Administration of sCD83 suppressed experimental FA. Pediatric FA patients have low serum sCD83 levels. Administration of sCD83 can alleviate experimental FA via suppression of aberrant Th2 polarization.
Collapse
Affiliation(s)
- Wenyue Song
- Departments of Pediatrics, Obstetrics and Gynecology, Jiaozuo Women and Children Hospital, 158 Minzhu Zhong Road, Jiaozuo, 454150, China
| | - Hongfen Li
- Departments of Pediatrics, Obstetrics and Gynecology, Jiaozuo Women and Children Hospital, 158 Minzhu Zhong Road, Jiaozuo, 454150, China
| | - Bingkun Jia
- Departments of Pediatrics, Obstetrics and Gynecology, Jiaozuo Women and Children Hospital, 158 Minzhu Zhong Road, Jiaozuo, 454150, China
| | - Zhenxi Wang
- Departments of Pediatrics, Obstetrics and Gynecology, Jiaozuo Women and Children Hospital, 158 Minzhu Zhong Road, Jiaozuo, 454150, China
| | - Qingsheng Liu
- Departments of Pediatrics, Obstetrics and Gynecology, Jiaozuo Women and Children Hospital, 158 Minzhu Zhong Road, Jiaozuo, 454150, China
| | - Guangping Yang
- Departments of Pediatrics, Obstetrics and Gynecology, Jiaozuo Women and Children Hospital, 158 Minzhu Zhong Road, Jiaozuo, 454150, China
| | - Xiaorong Li
- Departments of Pediatrics, Obstetrics and Gynecology, Jiaozuo Women and Children Hospital, 158 Minzhu Zhong Road, Jiaozuo, 454150, China.
| |
Collapse
|
35
|
Hyaluronic acid binding to CD44S is indiscriminate of molecular weight. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1862:183348. [PMID: 32428448 DOI: 10.1016/j.bbamem.2020.183348] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/09/2020] [Accepted: 05/07/2020] [Indexed: 01/06/2023]
Abstract
The ubiquitous presence of hyaluronic acid (HA) in the extracellular matrix (ECM) of both healthy and diseased tissues underscores its importance in human physiology. Previous studies suggest that HA can be used as a probe to qualitatively monitor cell surface levels of CD44 and other important HA receptors; however, these studies use mixtures of HA at various molecular weights. Using fluorescently labeled HA, we evaluated the apparent differences of low (25 kilodalton) and high (700 kilodalton) molecular weight HA interacting with breast cancer cell lines of varying levels of CD44. Our results confirm that CD44 expression and the apparent level of HA interaction correlates with molecular weight. Importantly, we show that HA only binds a small fraction of the major CD44 isoform, CD44S, on cell surfaces and that CD44S interactions account for <50% of the total HA bound to cell surfaces. Although increased fluorescence level correlates with higher molecular weight of HA, this appears to be an artifact of chain length and not a result of multivalent binding between HA and CD44S. Accordingly, we verify that HA binding characteristics of cell surfaces is similar to previous artificial membrane models which proposed that HA anchors to CD44S and forms a non-binding corona of HA that extends beyond the surface.
Collapse
|
36
|
Grosche L, Knippertz I, König C, Royzman D, Wild AB, Zinser E, Sticht H, Muller YA, Steinkasserer A, Lechmann M. The CD83 Molecule - An Important Immune Checkpoint. Front Immunol 2020; 11:721. [PMID: 32362900 PMCID: PMC7181454 DOI: 10.3389/fimmu.2020.00721] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022] Open
Abstract
The CD83 molecule has been identified to be expressed on numerous activated immune cells, including B and T lymphocytes, monocytes, dendritic cells, microglia, and neutrophils. Both isoforms of CD83, the membrane-bound as well as its soluble form are topic of intensive research investigations. Several studies revealed that CD83 is not a typical co-stimulatory molecule, but rather plays a critical role in controlling and resolving immune responses. Moreover, CD83 is an essential factor during the differentiation of T and B lymphocytes, and the development and maintenance of tolerance. The identification of its interaction partners as well as signaling pathways have been an enigma for the last decades. Here, we report the latest data on the expression, structure, and the signaling partners of CD83. In addition, we review the regulatory functions of CD83, including its striking modulatory potential to maintain the balance between tolerance versus inflammation during homeostasis or pathologies. These immunomodulatory properties of CD83 emphasize its exceptional therapeutic potential, which has been documented in specific preclinical disease models.
Collapse
Affiliation(s)
- Linda Grosche
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ilka Knippertz
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christina König
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas B. Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Yves A. Muller
- Division of Biotechnology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Lechmann
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
37
|
Wild AB, Krzyzak L, Peckert K, Stich L, Kuhnt C, Butterhof A, Seitz C, Mattner J, Grüner N, Gänsbauer M, Purtak M, Soulat D, Winkler TH, Nitschke L, Zinser E, Steinkasserer A. CD83 orchestrates immunity toward self and non-self in dendritic cells. JCI Insight 2019; 4:e126246. [PMID: 31527313 PMCID: PMC6824307 DOI: 10.1172/jci.insight.126246] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 09/04/2019] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) are crucial to balance protective immunity and autoimmune inflammatory processes. Expression of CD83 is a well-established marker for mature DCs, although its physiological role is still not completely understood. Using a DC-specific CD83-conditional KO (CD83ΔDC) mouse, we provide new insights into the function of CD83 within this cell type. Interestingly, CD83-deficient DCs produced drastically increased IL-2 levels and displayed higher expression of the costimulatory molecules CD25 and OX40L, which causes superior induction of antigen-specific T cell responses and compromises Treg suppressive functions. This also directly translates into accelerated immune responses in vivo. Upon Salmonella typhimurium and Listeria monocytogenes infection, CD83ΔDC mice cleared both pathogens more efficiently, and CD83-deficient DCs expressed increased IL-12 levels after bacterial encounter. Using the experimental autoimmune encephalomyelitis model, autoimmune inflammation was dramatically aggravated in CD83ΔDC mice while resolution of inflammation was strongly reduced. This phenotype was associated with increased cell influx into the CNS accompanied by elevated Th17 cell numbers. Concomitantly, CD83ΔDC mice had reduced Treg numbers in peripheral lymphoid organs. In summary, we show that CD83 ablation on DCs results in enhanced immune responses by dysregulating tolerance mechanisms and thereby impairing resolution of inflammation, which also demonstrates high clinical relevance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jochen Mattner
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Niklas Grüner
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maximilian Gänsbauer
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Purtak
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Didier Soulat
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas H. Winkler
- Division of Genetics, Department of Biology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | |
Collapse
|
38
|
Huo S, Zhang J, Liang S, Wu F, Zuo Y, Cui D, Zhang Y, Zhong Z, Zhong F. Membrane-bound and soluble porcine CD83 functions antithetically in T cell activation and dendritic cell differentiation in vitro. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 99:103398. [PMID: 31121186 DOI: 10.1016/j.dci.2019.103398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 06/09/2023]
Abstract
Emerging evidence suggests that CD83, a dendritic cells (DCs) maturation marker in humans and mice, may prossess immunomodulatory capacities. Although porcine CD83 shares ∼75% sequence homology with its human counterpart, whether it functions as an immunoregulatory molecule remains unknown. To investigate porcine CD83 function, we deleted it in porcine DCs by RNA intereference. Results show that membrane-bound CD83 (mCD83) promotes DC-mediated T cell proliferation and cytokine production, thus confirming its immunoregulatory capacity. Intriguingly, porcine soluble CD83 (sCD83) treatment instead led to inhibition of DC-mediated T cell activation. Moreover, porcine sCD83 also inhibited differentiation of prepheral blood mononuclear cells (PBMCs) into DCs. These results collectively indicate that in addition to being a DC maturation maker, both membrane bound and souble porcine CD83 serve as immunoregulatory molecules with opposite effects on DC-mediated T cell activation and DC differentiation.
Collapse
Affiliation(s)
- Shanshan Huo
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Jianlou Zhang
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Shuang Liang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Fengyang Wu
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Yuzhu Zuo
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Dan Cui
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Yonghong Zhang
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Zhenyu Zhong
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Fei Zhong
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China.
| |
Collapse
|
39
|
Mölzer C, Shankar SP, Griffith M, Islam MM, Forrester JV, Kuffová L. Activation of dendritic cells by crosslinked collagen hydrogels (artificial corneas) varies with their composition. J Tissue Eng Regen Med 2019; 13:1528-1543. [PMID: 31144475 DOI: 10.1002/term.2903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 05/01/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022]
Abstract
Activated T cells are known to promote fibrosis, a major complication limiting the range of polymeric hydrogels as artificial corneal implants. As T cells are activated by dendritic cells (DC), minimally activating hydrogels would be optimal. In this study, we evaluated the ability of a series of engineered (manufactured/fabricated) and natural collagen matrices to either activate DC or conversely induce DC apoptosis in vitro. Bone marrow DC were cultured on a series of singly and doubly crosslinked hydrogels (made from recombinant human collagen III [RHCIII] or collagen mimetic peptide [CMP]) or on natural collagen-containing matrices, MatrigelTM and de-cellularised mouse corneal stroma. DC surface expression of major histocompatibility complex Class II and CD86 as well as apoptosis markers were examined. Natural matrices induced low levels of DC activation and maintained a "tolerogenic" phenotype. The same applied to singly crosslinked CMP-PEG gels. RHCIII gels singly crosslinked using either N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide with the coinitiator N-hydroxy succinimide (EDC-NHS) or N-cyclohexyl-N-(2-morpholinoethyl)carbodiimide metho-p-toulenesulfonate with NHS (CMC-NHS) induced varying levels of DC activation. In contrast, however, RHCIII hydrogels incorporating an additional polymeric network of 2-methacryloyloxyethyl phosphorylcholine did not activate DC but instead induced DC apoptosis, a phenomenon observed in natural matrices. This correlated with increased DC expression of leukocyte-associated immunoglobulin-like receptor-1. Despite low immunogenic potential, viable tolerogenic DC migrated into and through both natural and manufactured RHCIII gels. These data show that the immunogenic potential of RHCIII gels varies with the nature and composition of the gel. Preclinical evaluation of hydrogel immunogenic/fibrogenic potential is recommended.
Collapse
Affiliation(s)
- Christine Mölzer
- School of Medicine and Dentistry, Section of Immunology, Inflammation and Infection, Institute of Medical Sciences, Division of Applied Medicine, University of Aberdeen, Aberdeen, UK
| | - Sucharita P Shankar
- School of Medicine and Dentistry, Section of Immunology, Inflammation and Infection, Institute of Medical Sciences, Division of Applied Medicine, University of Aberdeen, Aberdeen, UK
| | - May Griffith
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, QC, Canada
| | - Mirazul M Islam
- Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,Schepens Eye Research Institute and Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - John V Forrester
- School of Medicine and Dentistry, Section of Immunology, Inflammation and Infection, Institute of Medical Sciences, Division of Applied Medicine, University of Aberdeen, Aberdeen, UK
| | - Lucia Kuffová
- School of Medicine and Dentistry, Section of Immunology, Inflammation and Infection, Institute of Medical Sciences, Division of Applied Medicine, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
40
|
Zinser E, Naumann R, Wild AB, Michalski J, Deinzer A, Stich L, Kuhnt C, Steinkasserer A, Knippertz I. Endogenous Expression of the Human CD83 Attenuates EAE Symptoms in Humanized Transgenic Mice and Increases the Activity of Regulatory T Cells. Front Immunol 2019; 10:1442. [PMID: 31293592 PMCID: PMC6603205 DOI: 10.3389/fimmu.2019.01442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/07/2019] [Indexed: 12/17/2022] Open
Abstract
The CD83 is a type I membrane protein and part of the immunoglobulin superfamily of receptors. CD83 is involved in the regulation of antigen presentation and dendritic cell dependent allogeneic T cell proliferation. A soluble form of CD83 inhibits dendritic cell maturation and function. Furthermore, CD83 is expressed on activated B cells, T cells, and in particular on regulatory T cells. Previous studies on murine CD83 demonstrated this molecule to be involved in several immune-regulatory processes, comprising that CD83 plays a key role in the development und function of different immune cells. In order to get further insights into the function of the human CD83 and to provide preclinical tools to guide the function of CD83/sCD83 for therapeutic purposes we generated Bacterial Artificial Chromosomes (BAC) transgenic mice. BACs are excellent tools for manipulating large DNA fragments and are utilized to engineer transgenic mice by pronuclear injection. Two different founders of BAC transgenic mice expressing human CD83 (BAC-hCD83tg mice) were generated and were examined for the hCD83 expression on different immune cells as well as both the in vitro and in vivo role of human CD83 (hCD83) in health and disease. Here, we found the hCD83 molecule to be present on activated DCs, B cells and subtypes of CD4+ T cells. CD8+ T cells, on the other hand, showed almost no hCD83 expression. To address the function of hCD83, we performed in vitro mixed lymphocyte reactions (MLR) as well as suppression assays and we used the in vivo model of experimental autoimmune encephalomyelitis (EAE) comparing wild-type and hCD83-BAC mice. Results herein showed a clearly diminished capacity of hCD83-BAC-derived T cells to proliferate accompanied by an enhanced activation and suppressive activity of hCD83-BAC-derived Tregs. Furthermore, hCD83-BAC mice were found to recover faster from EAE-associated symptoms than wild-type mice, encouraging the relevance also of the hCD83 as a key molecule for the regulatory phenotype of Tregs in vitro and in vivo.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Humans
- Immunoglobulins/genetics
- Immunoglobulins/immunology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Transgenic
- Somatostatin-Secreting Cells/immunology
- Somatostatin-Secreting Cells/pathology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- CD83 Antigen
Collapse
Affiliation(s)
- Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ronald Naumann
- Transgenic Core Facility, Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andreas B. Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Michalski
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andrea Deinzer
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christine Kuhnt
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Ilka Knippertz
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
41
|
Li Z, Ju X, Silveira PA, Abadir E, Hsu WH, Hart DNJ, Clark GJ. CD83: Activation Marker for Antigen Presenting Cells and Its Therapeutic Potential. Front Immunol 2019; 10:1312. [PMID: 31231400 PMCID: PMC6568190 DOI: 10.3389/fimmu.2019.01312] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/23/2019] [Indexed: 12/17/2022] Open
Abstract
CD83 is a member of the immunoglobulin (Ig) superfamily and is expressed in membrane bound or soluble forms. Membrane CD83 (mCD83) can be detected on a variety of activated immune cells, although it is most highly and stably expressed by mature dendritic cells (DC). mCD83 regulates maturation, activation and homeostasis. Soluble CD83 (sCD83), which is elevated in the serum of patients with autoimmune disease and some hematological malignancies is reported to have an immune suppressive function. While CD83 is emerging as a promising immune modulator with therapeutic potential, some important aspects such as its ligand/s, intracellular signaling pathways and modulators of its expression are unclear. In this review we discuss the recent biological findings and the potential clinical value of CD83 based therapeutics in various conditions including autoimmune disease, graft-vs.-host disease, transplantation and hematological malignancies.
Collapse
Affiliation(s)
- Ziduo Li
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Xinsheng Ju
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Pablo A. Silveira
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Edward Abadir
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Wei-Hsun Hsu
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Derek N. J. Hart
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Georgina J. Clark
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
42
|
Aladily TN, Mansour A, Alsughayer A, Sughayer M, Medeiros LJ. The utility of CD83, fascin and CD23 in the differential diagnosis of primary mediastinal large B-cell lymphoma versus classic Hodgkin lymphoma. Ann Diagn Pathol 2019; 40:72-76. [PMID: 31075666 DOI: 10.1016/j.anndiagpath.2019.04.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 01/16/2023]
Abstract
Primary mediastinal large B-cell lymphoma (PMBL) and classic Hodgkin lymphoma (CHL) are the most common large cell lymphomas arising in the mediastinum and are thought to be closely related histogenetically. Although the distinction between PMBL and CHL is usually straightforward, in some cases it is challenging and rarely these neoplasms have intermediate features and qualify for the diagnosis of mediastinal gray zone lymphoma (GZL). CD83 and fascin are markers of CHL and CD23 is a marker of PMBL. In this study we assess the utility of this combination of these immunohistochemical markers to distinguish CHL from PMBL. We retrospectively collected cases of PMBL, CHL and GZL from three centers. Tissue sections were stained with CD83, fascin and CD23. CD83 was expressed in the neoplastic cells of 100% of CHL (22/22), 93% of GZL (16/18) and 41% of PMBL (9/22). Similarly, fascin was positive in the neoplastic cells of 100% of CHL (22/22), 86% of GZL (18/21) and 32% of PMBL (7/22). CD23 was positive in 95% of PMBL (21/22), 67% of GZL (12/18) and 9% of CHL (2/22). CD83 and fascin are sensitive markers for CHL but not specific whereas CD23 is sensitive for PMBL and uncommon in CHL. The GZL cases in this study had an intermediate immunophenotype, but the results were closer to CHL than PMBL. A large panel of immunohistochemical studies is recommended to distinguish CHL from PMBL entities and we suggest that CD83, fascin and CD23 add value to panels designed for this differential diagnosis.
Collapse
MESH Headings
- Antigens, CD/metabolism
- Biomarkers, Tumor/metabolism
- Carrier Proteins/metabolism
- Diagnosis, Differential
- Hodgkin Disease/diagnosis
- Hodgkin Disease/metabolism
- Hodgkin Disease/pathology
- Humans
- Immunoglobulins/metabolism
- Immunohistochemistry
- Immunophenotyping
- Lymphoma, Large B-Cell, Diffuse/diagnosis
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Mediastinal Neoplasms/diagnosis
- Mediastinal Neoplasms/metabolism
- Mediastinal Neoplasms/pathology
- Membrane Glycoproteins/metabolism
- Microfilament Proteins/metabolism
- Receptors, IgE/metabolism
- Retrospective Studies
- CD83 Antigen
Collapse
Affiliation(s)
- Tariq N Aladily
- Department of Pathology, The University of Jordan, Queen Rania St, Amman 11942, Jordan.
| | - Ahmad Mansour
- Department of Pathology, The University of Jordan, Queen Rania St, Amman 11942, Jordan
| | - Anas Alsughayer
- Department of Pathology, The University of Jordan, Queen Rania St, Amman 11942, Jordan
| | - Maher Sughayer
- Department of Pathology, King Hussein Cancer Center, Amman, Queen Rania St, Amman 11941, Jordan.
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe blvd, Houston, TX 77030, USA.
| |
Collapse
|
43
|
Royzman D, Andreev D, Stich L, Rauh M, Bäuerle T, Ellmann S, Boon L, Kindermann M, Peckert K, Bozec A, Schett G, Steinkasserer A, Zinser E. Soluble CD83 Triggers Resolution of Arthritis and Sustained Inflammation Control in IDO Dependent Manner. Front Immunol 2019; 10:633. [PMID: 31001257 PMCID: PMC6455294 DOI: 10.3389/fimmu.2019.00633] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Interference with autoimmune-mediated cytokine production is a key yet poorly developed approach to treat autoimmune and inflammatory diseases such as rheumatoid arthritis. Herein, we show that soluble CD83 (sCD83) enhances the resolution of autoimmune antigen-induced arthritis (AIA) by strongly reducing the expression levels of cytokines such as IL-17A, IFNγ, IL-6, and TNFα within the joints. Noteworthy, also the expression of RANKL, osteoclast differentiation, and joint destruction was significantly inhibited by sCD83. In addition, osteoclasts which were cultured in the presence of synovial T cells, derived from sCD83 treated AIA mice, showed a strongly reduced number of multinuclear large osteoclasts compared to mock controls. Enhanced resolution of arthritis by sCD83 was mechanistically based on IDO, since inhibition of IDO by 1-methyltryptophan completely abrogated sCD83 effects on AIA. Blocking experiments, using anti-TGF-β antibodies further revealed that also TGF-β is mechanistically involved in the sCD83 induced reduction of bone destruction and cartilage damage as well as enhanced resolution of inflammation. Resolution of arthritis was associated with increased numbers of regulatory T cells, which are induced in a sCD83-IDO-TGF-β dependent manner. Taken together, sCD83 represents an interesting approach for downregulating cytokine production, inducing regulatory T cells and inducing resolution of autoimmune arthritis.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/pharmacology
- Antigens, CD/immunology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Cytokines/immunology
- Female
- Immunoglobulins/immunology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/pathology
- Joints/immunology
- Joints/pathology
- Membrane Glycoproteins/immunology
- Mice
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Solubility
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/immunology
- Tryptophan/analogs & derivatives
- Tryptophan/pharmacology
- CD83 Antigen
Collapse
Affiliation(s)
- Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Darja Andreev
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stephan Ellmann
- Institute of Radiology, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Markus Kindermann
- Department of Internal Medicine 1, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katrin Peckert
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
44
|
Affiliation(s)
- Ralf Küppers
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Medical Faculty, Germany
| |
Collapse
|
45
|
Juhas U, Ryba-Stanisławowska M, Ławrynowicz U, Myśliwiec M, Myśliwska J. Putative loss of CD83 immunosuppressive activity in long-standing complication-free juvenile diabetic patients during disease progression. Immunol Res 2019; 67:70-76. [PMID: 30937729 DOI: 10.1007/s12026-019-09074-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The CD83 molecule is a known marker of dendritic cell differentiation process, and its soluble form (sCD83) exerts immunosuppressive functions. In our research, we examined whether the sCD83 plasma concentration is impaired in DM1 children and if the expected changes are in line with the disturbed process of monocyte's transformation into mCD83+ monocyte-derived cells. 28 newly diagnosed (ND-DM1) and 30 long-standing (LS-DM1) patients were enrolled into our study. We revealed that the examined cells show a high mCD83 expression level in ND-DM1, which was significantly downregulated by the TNF-α stimulation. The results were in line with those from healthy controls. We also observed that monocyte differentiation process into CD83+ cells was much defective in LS-DM1 children and the mCD83 expression level seems not to be controlled by TNF-α. Moreover, the sCD83 level was significantly decreased in plasma from LS-DM1 children and it was negatively related to HbA1c levels, while no correlations were observed between TNF-α plasma concentration or disease duration. Summarizing, our results suggest that reduced sCD83 levels may correspond with a poor metabolic control in LS-DM1 patients and therapeutic administration of this molecule may indicate a new therapy approach in the chronic phase of diabetes.
Collapse
Affiliation(s)
- Ulana Juhas
- Department of Medical Immunology, Laboratory of Experimental Immunology, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland.
| | - Monika Ryba-Stanisławowska
- Department of Medical Immunology, Laboratory of Experimental Immunology, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland
| | - Urszula Ławrynowicz
- Department of Medical Immunology, Laboratory of Experimental Immunology, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland
| | - Małgorzata Myśliwiec
- Department of Paediatrics, Diabetology and Endocrinology, Medical University of Gdańsk, Dębinki 7, 80-211, Gdańsk, Poland
| | - Jolanta Myśliwska
- Department of Medical Immunology, Laboratory of Experimental Immunology, Medical University of Gdańsk, Dębinki 1, 80-211, Gdańsk, Poland
| |
Collapse
|
46
|
Hu J, Shi B, Liu X, Jiang M, Yuan C, Jiang B, Song Y, Zeng Y, Wang G. The activation of Toll-like receptor 4 reverses tumor differentiation in human glioma U251 cells via Notch pathway. Int Immunopharmacol 2018; 64:33-41. [PMID: 30145468 DOI: 10.1016/j.intimp.2018.08.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 08/05/2018] [Accepted: 08/15/2018] [Indexed: 12/23/2022]
Abstract
Toll-like receptors (TLRs) are closely related to cancer. However, the mechanism for TLR regulation of cancer is not fully understood. Our previous studies demonstrated that toll-like receptor (TLR) 4 functions to maintain the un-differential stem cell phenotypes of human endothelial progenitor cells. In this study, we found that human glioma cells expressed several TLRs. The activation of TLR4 by LPS in glioma U251 cells induced the expression of cytokines, including IL-1β, IL-6, IL-8, and TNFα, suggesting the functional expression of TLR4. Nude mouse in vivo studies showed that LPS treatment promoted tumor growth, and decreased mouse survival. But LPS treatment did not promote tumor cell proliferation in vitro. Meanwhile, we found that LPS treatment down-regulated the expression of glial fibrillary acidic protein (GFAP), an important differentiation maker of glioma, at both mRNA and protein levels. TLR4 activation also down-regulated GFAP in glioma Hs683 cells. LPS did not induce the activation of MAPKs, but induced the activation of NF-κB. However, pharmacological inhibition of NF-κB signaling did not reverse the down-regulation of GFAP. Furthermore, we found that LPS induced the activation of Notch pathway, which was MyD88-dependent, and Notch inhibition reversed the down-regulation of GFAP. In addition, LPS treatment up-regulated stem cell makers, including CD34 and CD133. Taken together, these results suggested that in human glioma U251 cells, TLR4 functions to reverse tumor differentiation, and it may be a target for glioma prevention and therapy.
Collapse
Affiliation(s)
- Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China.
| | - Bizhi Shi
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Chuang Yuan
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Binyuan Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Yinghui Song
- Department of Oncology, Changsha Central Hospital, Changsha, Hunan 410004, China; Changsha Cancer Institute, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Yanhua Zeng
- Department of Oncology, Changsha Central Hospital, Changsha, Hunan 410004, China; Changsha Cancer Institute, Changsha Central Hospital, Changsha, Hunan 410004, China
| | - Guihua Wang
- Department of Oncology, Changsha Central Hospital, Changsha, Hunan 410004, China; Changsha Cancer Institute, Changsha Central Hospital, Changsha, Hunan 410004, China.
| |
Collapse
|
47
|
Chen X, Bai J, Liu X, Song Z, Zhang Q, Wang X, Jiang P. Nsp1α of Porcine Reproductive and Respiratory Syndrome Virus Strain BB0907 Impairs the Function of Monocyte-Derived Dendritic Cells via the Release of Soluble CD83. J Virol 2018; 92:e00366-18. [PMID: 29793955 PMCID: PMC6052304 DOI: 10.1128/jvi.00366-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/08/2018] [Indexed: 12/21/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV), a virulent pathogen of swine, suppresses the innate immune response and induces persistent infection. One mechanism used by viruses to evade the immune system is to cripple the antigen-processing machinery in monocyte-derived dendritic cells (MoDCs). In this study, we show that MoDCs infected by PRRSV express lower levels of the major histocompatibility complex (MHC)-peptide complex proteins TAP1 and ERp57 and are impaired in their ability to stimulate T cell proliferation and increase their production of CD83. Neutralization of sCD83 removes the inhibitory effects of PRRSV on MoDCs. When MoDCs are incubated with exogenously added sCD83 protein, TAP1 and ERp57 expression decreases and T lymphocyte activation is impaired. PRRSV nonstructural protein 1α (Nsp1α) enhances CD83 promoter activity. Mutations in the ZF domain of Nsp1α abolish its ability to activate the CD83 promoter. We generated recombinant PRRSVs with mutations in Nsp1α and the corresponding repaired PRRSVs. Viruses with Nsp1α mutations did not decrease levels of TAP1 and ERp57, impair the ability of MoDCs to stimulate T cell proliferation, or increase levels of sCD83. We show that the ZF domain of Nsp1α stimulates the secretion of CD83, which in turn inhibits MoDC function. Our study provides new insights into the mechanisms of immune suppression by PRRSV.IMPORTANCE PRRSV has a severe impact on the swine industry throughout the world. Understanding the mechanisms by which PRRSV infection suppresses the immune system is essential for a robust and sustainable swine industry. Here, we demonstrated that PRRSV infection manipulates MoDCs by interfering with their ability to produce proteins in the MHC-peptide complex. The virus also impairs the ability of MoDCs to stimulate cell proliferation, due in large part to the enhanced release of soluble CD83 from PRRSV-infected MoDCs. The viral nonstructural protein 1 (Nsp1) is responsible for upregulating CD83 promoter activity. Amino acids in the ZF domain of Nsp1α (L5-2A, rG45A, G48A, and L61-6A) are essential for CD83 promoter activation. Viruses with mutations at these sites no longer inhibit MoDC-mediated T cell proliferation. These findings provide novel insights into the mechanism by which the adaptive immune response is suppressed during PRRSV infection.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xuewei Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhongbao Song
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qiaoya Zhang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xianwei Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
48
|
Lin W, Buscher K, Wang B, Fan Z, Song N, Li P, Yue Y, Li B, Li C, Bi H. Soluble CD83 Alleviates Experimental Autoimmune Uveitis by Inhibiting Filamentous Actin-Dependent Calcium Release in Dendritic Cells. Front Immunol 2018; 9:1567. [PMID: 30050530 PMCID: PMC6052908 DOI: 10.3389/fimmu.2018.01567] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/25/2018] [Indexed: 01/03/2023] Open
Abstract
Soluble CD83 (sCD83) is the extracellular domain of the membrane-bound CD83 molecule, and known for its immunoregulatory functions. Whether and how sCD83 participates in the pathogenesis of uveitis, a serious inflammatory disease of the eye that can cause visual disability and blindness, is unknown. By flow cytometry and imaging studies, we show that sCD83 alleviates experimental autoimmune uveitis (EAU) through a novel mechanism. During onset and recovery of EAU, the level of sCD83 rises in the serum and aqueous humor, and CD83+ leukocytes infiltrate the inflamed eye. Systemic or topical application of sCD83 exerts a protective effect by decreasing inflammatory cytokine expression, reducing ocular and splenic leukocyte including CD4+ T cells and dendritic cells (DCs). Mechanistically, sCD83 induces tolerogenic DCs by decreasing the synaptic expression of co-stimulatory molecules and hampering the calcium response in DCs. These changes are caused by a disruption of the cytoskeletal rearrangements at the DC–T cell contact zone, leading to altered localization of calcium microdomains and suppressed T-cell activation. Thus, the ability of sCD83 to modulate DC-mediated inflammation in the eye could be harnessed to develop new immunosuppressive therapeutics for autoimmune uveitis.
Collapse
Affiliation(s)
- Wei Lin
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, China.,Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Immunology, Shanghai Medical School, Fudan University, Shanghai, China
| | - Konrad Buscher
- Department of Nephrology and Rheumatology, University Hospital Muenster, Münster, Germany.,Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - Beibei Wang
- Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhichao Fan
- Division of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States
| | - Nannan Song
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Peng Li
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Yingying Yue
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Bingqing Li
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Cuiling Li
- Institute of Basic Medicine, Shandong Academy of Medical Sciences, Jinan, China
| | - Hongsheng Bi
- Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
49
|
Takheaw N, Laopajon W, Surinkaew S, Khummuang S, Pata S, Kasinrerk W. Ligation of Na, K ATPase β3 subunit on monocytes by a specific monoclonal antibody mediates T cell hypofunction. PLoS One 2018; 13:e0199717. [PMID: 29940031 PMCID: PMC6016913 DOI: 10.1371/journal.pone.0199717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/12/2018] [Indexed: 12/20/2022] Open
Abstract
T cells play a crucial role in orchestrating body immune responses. T cell hyperfunction, however, leads to inflammation and induction of autoimmune diseases. Understanding of T cell regulation mechanisms and successful modulation of T cell responses is beneficial in treatment of disease associated to T cell hyperresponsiveness. Our previous study indicated that monoclonal antibody (mAb) P-3E10, a mAb to Na, K ATPase β3 subunit, inhibited anti-CD3-induced PBMC proliferation. In the current study, we further investigated the mechanism of mAb P-3E10 in the induction of T cell hypofunction. We demonstrated that mAb P-3E10 decreased T cell proliferation and Th1, Th2 and Th17 cytokine production. Monocytes were the cells playing a key role in mediation of mAb P-3E10 induced T cell hypofunction. The inhibition of T cell activation by mAb P-3E10 required cell contact between monocytes and T cells. The mAb P-3E10 induced the down-expression level of MHC class II and CD86 and increased IL-6, IL-10 and TNF-α production of monocytes. We concluded that ligation of the Na, K ATPase β3 subunit on monocytes by mAb P-3E10 arbitrated T cell hypofunction. This mAb might be a promising novel immunotherapeutic antibody for the treatment of hyperresponsive T cell associated diseases.
Collapse
Affiliation(s)
- Nuchjira Takheaw
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Witida Laopajon
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Sirirat Surinkaew
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Saichit Khummuang
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Supansa Pata
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Watchara Kasinrerk
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
50
|
Doebbeler M, Koenig C, Krzyzak L, Seitz C, Wild A, Ulas T, Baßler K, Kopelyanskiy D, Butterhof A, Kuhnt C, Kreiser S, Stich L, Zinser E, Knippertz I, Wirtz S, Riegel C, Hoffmann P, Edinger M, Nitschke L, Winkler T, Schultze JL, Steinkasserer A, Lechmann M. CD83 expression is essential for Treg cell differentiation and stability. JCI Insight 2018; 3:99712. [PMID: 29875316 PMCID: PMC6124443 DOI: 10.1172/jci.insight.99712] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/01/2018] [Indexed: 12/21/2022] Open
Abstract
Foxp3-positive regulatory T cells (Tregs) are crucial for the maintenance of immune homeostasis and keep immune responses in check. Upon activation, Tregs are transferred into an effector state expressing transcripts essential for their suppressive activity, migration, and survival. However, it is not completely understood how different intrinsic and environmental factors control differentiation. Here, we present for the first time to our knowledge data suggesting that Treg-intrinsic expression of CD83 is essential for Treg differentiation upon activation. Interestingly, mice with Treg-intrinsic CD83 deficiency are characterized by a proinflammatory phenotype. Furthermore, the loss of CD83 expression by Tregs leads to the downregulation of Treg-specific differentiation markers and the induction of an inflammatory profile. In addition, Treg-specific conditional knockout mice showed aggravated autoimmunity and an impaired resolution of inflammation. Altogether, our results show that CD83 expression in Tregs is an essential factor for the development and function of effector Tregs upon activation. Since Tregs play a crucial role in the maintenance of immune tolerance and thus prevention of autoimmune disorders, our findings are also clinically relevant.
Collapse
Affiliation(s)
- Marina Doebbeler
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Christina Koenig
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Lena Krzyzak
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Christine Seitz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Andreas Wild
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Thomas Ulas
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Kevin Baßler
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Dmitry Kopelyanskiy
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Alina Butterhof
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Christine Kuhnt
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Simon Kreiser
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Ilka Knippertz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - Christin Riegel
- Department of Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| | - Petra Hoffmann
- Department of Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Edinger
- Department of Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Winkler
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Joachim L. Schultze
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Matthias Lechmann
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|