1
|
Shu J, Wang K, Liu Y, Zhang J, Ding X, Sun H, Wu J, Huang B, Qiu J, Sheng H, Lu L. Trichosanthin alleviates streptozotocin-induced type 1 diabetes mellitus in mice by regulating the balance between bone marrow-derived IL6 + and IL10 + MDSCs. Heliyon 2024; 10:e22907. [PMID: 38187307 PMCID: PMC10770427 DOI: 10.1016/j.heliyon.2023.e22907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 11/15/2023] [Accepted: 11/22/2023] [Indexed: 01/09/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) occupy a pivotal role in the intricate pathogenesis of the autoimmune disorder, Type 1 diabetes mellitus (T1DM). Since our previous work demonstrated that trichosanthin (TCS), an active compound of Chinese herb medicine Tian Hua Fen, regulated immune response, we aimed to clarify the efficacy and molecular mechanism of TCS in the treatment of T1DM. To this end, T1DM mouse model was established by streptozotocin (STZ) induction. The mice were randomly divided into normal control group (Ctl), T1DM group (STZ), TCS treated diabetic group (STZ + TCS) and insulin-treated diabetic group (STZ + insulin). Our comprehensive evaluation encompassed variables such as blood glucose, glycosylated hemoglobin, body weight, pertinent biochemical markers, pancreatic histopathology, and the distribution of immune cell populations. Furthermore, we meticulously isolated MDSCs from the bone marrow of T1DM mice, probing into the expressions of genes pertaining to the advanced glycation end product receptor (RAGE)/NF-κB signaling pathway through RT-qPCR. Evidently, TCS exhibited a substantial capacity to effectively counteract the T1DM-induced elevation in random blood glucose, glycosylated hemoglobin, and IL-6 levels in plasma. Pathological scrutiny underscored the ability of TCS to mitigate the damage incurred by islets. Intriguingly, TCS interventions engendered a reduction in the proportion of MDSCs within the bone marrow, particularly within the IL-6+ MDSC subset. In contrast, IL-10+ MDSCs exhibited an elevation following TCS treatment. Moreover, we observed a significant down-regulation of relative mRNA of pro-inflammatory genes, including arginase 1 (Arg1), inducible nitric oxide synthase (iNOS), RAGE and NF-κB, within MDSCs due to the influence of TCS. It decreases total MDSCs and regulates the balance between IL-6+ and IL-10+ MDSCs thus alleviating the symptoms of T1DM. TCS also down-regulates the RAGE/NF-κB signaling pathway, making it a promising alternative therapeutic treatment for T1DM. Collectively, our study offered novel insights into the underlying mechanism by which TCS serves as a promising therapeutic intervention for T1DM.
Collapse
Affiliation(s)
- Jie Shu
- Department of Clinical Laboratory, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai, 200336, China
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 Chong Qing South Road, 200025, China
| | - Kefan Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 Chong Qing South Road, 200025, China
| | - Yuting Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 Chong Qing South Road, 200025, China
| | - Jie Zhang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 Chong Qing South Road, 200025, China
| | - Xuping Ding
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 Chong Qing South Road, 200025, China
| | - Hanxiao Sun
- Department of Clinical Laboratory, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai, 200336, China
| | - Jiaoxiang Wu
- Department of Clinical Laboratory, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai, 200336, China
| | - Biao Huang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Ju Qiu
- The Key Laboratory of Stem Cell Biology, Shanghai Institutes of Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huiming Sheng
- Department of Clinical Laboratory, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xian Xia Road, Shanghai, 200336, China
| | - Liming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 Chong Qing South Road, 200025, China
| |
Collapse
|
2
|
Liu T, Rosek A, Gonzalez De Los Santos F, Phan SH. Detection of myeloid-derived suppressor cells by flow cytometry. Methods Cell Biol 2023; 184:1-15. [PMID: 38555150 DOI: 10.1016/bs.mcb.2023.06.006] [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] [Indexed: 04/02/2024]
Abstract
Recently discovered heterogeneous myeloid-derived suppressor cells (MDSCs) are some of the most discussed immunosuppressive cells in contemporary immunology, especially in the tumor microenvironment, and are defined primarily by their T cell immunosuppressive function. The importance of these cells extend to other chronic pathological conditions as well, including chronic infection, inflammation, and tissue remodeling. In many of these conditions, their accumulation/expansion correlates with disease progression, poor prognosis, and reduced survival, which highlights the potential of how these cells may be used in a clinical setting as both prognostic factor and therapeutic target. In healthy individuals, these cells are usually not present in the circulation. Therefore, monitoring this cell population is of potential clinical significance, and utility in basic research. However, these cells have a complex phenotype without one single marker of sufficient specificity for their identification. Flow cytometry is a powerful tool allowing multi-parameter analysis of heterogeneous cell populations, which makes it ideally suitable for the complex phenotypic analysis essential for identification and enumeration of circulating MDSCs. This approach has the potential to provide a novel clinically useful tool for assessment of prognosis and treatment outcomes. The protocol in this chapter describes a flow cytometric analysis to identify and quantify MDSCs from human or mouse whole blood leukocytes and peripheral blood mononuclear cells, as well as a single cell suspension from solid tissue, by using multicolor fluorescence-conjugated antibodies against their surface markers.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States.
| | - Alyssa Rosek
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Sem H Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
3
|
BORAL B, TUNCER İ, KİBAR F, ÇETİNER S, BADAK SÖ, SALMAN E, KOÇ E, ERKEN E, YAMAN A. CD39 expression on immune cells predicts methotrexate response in rheumatoid arthritis patients. Turk J Med Sci 2023; 53:1075-1083. [PMID: 38813034 PMCID: PMC10763742 DOI: 10.55730/1300-0144.5672] [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: 04/12/2023] [Revised: 10/26/2023] [Accepted: 09/09/2023] [Indexed: 05/31/2024] Open
Abstract
Background/aim Rheumatoid arthritis (RA) is a chronic inflammatory disease affecting mostly small joints, such as hand and foot joints symmetrically with irreversible joint destruction. In this study, the relationship between CD39 expression and the treatment response of RA patients was examined to investigate its potential as a biomarker that demonstrates treatment response. Materials and methods This study included 77 RA patients and 40 healthy controls (HC). The RA patients were divided into 2 groups based on their response to RA treatment, those with a good response to methotrexate (MTX) monotherapy and those with an inadequate response based on the American College of Rheumatology and the European League Against Rheumatism response criteria. Various immunological parameters and Disease Activity Score in 28 Joints (DAS28) were examined between the groups using the Student's t-test. Results The monocytic myeloid-derived suppressor cell (M-MDSC) percentage was higher in the RA patient group versus the HC group. The CD39 expression in the T lymphocytes were higher in patients that responded well to the MTX compared to those showing inadequate response. Additionally, s negative correlation was found between the DAS28 and CD39 in the T cells. Conclusion The results showed that the improvement in treatment response to the therapy in RA patients could be because of the enhancement in the CD39/adenosine (ADO) pathway. Therefore, therapies targeting the CD39/ADO pathway in T cells may improve RA treatments.
Collapse
Affiliation(s)
- Barış BORAL
- Department of Immunology, Adana Health Practice and Research Center, University of Health Sciences, Adana,
Turkiye
| | - İbrahim TUNCER
- Department of Immunology, Prof. Dr. Cemil Taşcıoğlu City Hospital, University of Health Sciences, İstanbul,
Turkiye
| | - Filiz KİBAR
- Department of Medical Microbiology, Faculty of Medicine, Çukurova University, Adana,
Turkiye
| | - Salih ÇETİNER
- Department of Immunology, Faculty of Medicine, Çukurova University, Adana,
Turkiye
| | - Suade Özlem BADAK
- Division of Rheumatology, Department of Internal Medicine, Adana City Training and Research Hospital, Adana
Turkiye
| | - Emrah SALMAN
- Department of Immunology, Ankara City Hospital, University of Health Sciences, Ankara,
Turkiye
| | - Emrah KOÇ
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Çukurova University, Adana,
Turkiye
| | - Eren ERKEN
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Çukurova University, Adana,
Turkiye
| | - Akgün YAMAN
- Department of Immunology, Adana Health Practice and Research Center, University of Health Sciences, Adana,
Turkiye
| |
Collapse
|
4
|
Varela VA, da Silva Heinen LB, Marti LC, Caraciolo VB, Datoguia TS, Amano MT, Pereira WO. In vitro differentiation of myeloid suppressor cells (MDSC-like) from an immature myelomonocytic precursor THP-1. J Immunol Methods 2023; 515:113441. [PMID: 36848984 DOI: 10.1016/j.jim.2023.113441] [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: 11/21/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/27/2023]
Abstract
BACKGROUND Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population with a potent suppressor profile that regulates immune responses. These cells are one of the main components of the microenvironment of several diseases, including solid and hematologic tumors, autoimmunities, and chronic inflammation. However, their wide use in studies is limited due to they comprehend a rare population, which is difficult to isolate, expand, differentiate, and maintain in culture. Additionally, this population has a complex phenotypic and functional characterization. OBJECTIVE To develop a protocol for the in vitro production of MDSC-like population from the differentiation of the immature myeloid cell line THP-1. METHODS We stimulated THP-1 with G-CSF (100 ng/mL) and IL-4 (20 ng/mL) for seven days to differentiate into the MDSC-like profile. At the end of the protocol, we characterized these cells phenotypically and functionally by immunophenotyping, gene expression analysis, cytokine release dosage, lymphocyte proliferation, and NK-mediated killing essays. RESULTS We differentiate THP-1 cells in an MDSC-like population, named THP1-MDSC-like, which presented immunophenotyping and gene expression profiles compatible with that described in the literature. Furthermore, we verified that this phenotypic and functional differentiation did not deviate to a macrophage profile of M1 or M2. These THP1-MDSC-like cells secreted several immunoregulatory cytokines into the microenvironment, consistent with the suppressor profile related to MDSC. In addition, the supernatant of these cells decreased the proliferation of activated lymphocytes and impaired the apoptosis of leukemic cells induced by NK cells. CONCLUSIONS We developed an effective protocol for MDSC in vitro production from the differentiation of the immature myeloid cell line THP-1 induced by G-CSF and IL-4. Furthermore, we demonstrated that THP1-MDSC-like suppressor cells contribute to the immune escape of AML cells. Potentially, these THP1-MDSC-like cells can be applied on a large-scale platform, thus being able to impact the course of several studies and models such as cancer, immunodeficiencies, autoimmunity, and chronic inflammation.
Collapse
Affiliation(s)
- Vanessa Araújo Varela
- Faculdade Israelita de Ciências da Saúde Albert Einstein (FICSAE), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Luciana Cavalheiro Marti
- Faculdade Israelita de Ciências da Saúde Albert Einstein (FICSAE), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Victória Bulcão Caraciolo
- Faculdade Israelita de Ciências da Saúde Albert Einstein (FICSAE), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Tarcila Santos Datoguia
- Faculdade Israelita de Ciências da Saúde Albert Einstein (FICSAE), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Mariane Tami Amano
- Hospital Sírio Libanês, São Paulo, SP, Brazil; Department of Clinical and Experimental Oncology, Federal University of São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Welbert Oliveira Pereira
- Faculdade Israelita de Ciências da Saúde Albert Einstein (FICSAE), Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
5
|
Torres-Ruiz J, Absalón-Aguilar A, Reyes-Islas JA, Cassiano-Quezada F, Mejía-Domínguez NR, Pérez-Fragoso A, Maravillas-Montero JL, Núñez-Álvarez C, Juárez-Vega G, Culebro-Bermejo A, Gómez-Martín D. Peripheral expansion of myeloid-derived suppressor cells is related to disease activity and damage accrual in inflammatory myopathies. Rheumatology (Oxford) 2023; 62:775-784. [PMID: 35766810 DOI: 10.1093/rheumatology/keac374] [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: 03/29/2022] [Revised: 05/26/2022] [Accepted: 06/18/2022] [Indexed: 02/04/2023] Open
Abstract
OBJECTIVE To assess the proportion of myeloid-derived suppressor cells (MDSCs), their expression of arginase-1 and programmed cell death ligand 1 (PD-L1) and their relationship with the clinical phenotype of patients with idiopathic inflammatory myopathies (IIMs). METHODS We recruited 37 IIM adult patients and 10 healthy donors in Mexico City. We evaluated their clinical features, the proportion of MDSCs and their expression of PD-L1 and arginase-1 by flow cytometry. Polymorphonuclear (PMN)-MDSCs were defined as CD33dim, CD11b+ and CD66b+ while monocytic (M)-MDSCs were CD33+, CD11b+, HLA-DR- and CD14+. Serum cytokines were analysed with a multiplex assay. We compared the quantitative variables with the Kruskal-Wallis and Mann-Whitney U tests and assessed correlations with Spearman's ρ. RESULTS Most patients had dermatomyositis [n = 30 (81.0%)]. IIM patients had a peripheral expansion of PMN-MDSCs and M-MDSCs with an enhanced expression of arginase-1 and PD-L1. Patients with active disease had a decreased percentage {median 1.75% [interquartile range (IQR) 0.31-5.50 vs 10.71 [3.16-15.58], P = 0.011} of M-MDSCs and a higher absolute number of PD-L1+ M-MDSCs [median 23.21 cells/mm3 (IQR 11.16-148.9) vs 5.95 (4.66-102.7), P = 0.046] with increased expression of PD-L1 [median 3136 arbitrary units (IQR 2258-4992) vs 1961 (1885-2335), P = 0.038]. PD-L1 expression in PMN-MDSCs correlated with the visual analogue scale of pulmonary disease activity (r = 0.34, P = 0.040) and damage (r = 0.36, P = 0.031), serum IL-5 (r = 0.55, P = 0.003), IL-6 (r = 0.46, P = 0.003), IL-8 (r = 0.53, P = 0.018), IL-10 (r = 0.48, P = 0.005) and GM-CSF (r = 0.48, P = 0.012). M-MDSCs negatively correlated with the skeletal Myositis Intention to Treat Index (r = -0.34, P = 0.038) and positively with IL-6 (r = 0.40, P = 0.045). CONCLUSION MDSCs expressing arginase-1 and PD-L1 are expanded in IIM and correlate with disease activity, damage accrual and serum cytokines.
Collapse
Affiliation(s)
- Jiram Torres-Ruiz
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Abdiel Absalón-Aguilar
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Juan Alberto Reyes-Islas
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Fabiola Cassiano-Quezada
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Nancy R Mejía-Domínguez
- Red de Apoyo a la Investigación, Coordinacion de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alfredo Pérez-Fragoso
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - José Luis Maravillas-Montero
- Red de Apoyo a la Investigación, Coordinacion de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Carlos Núñez-Álvarez
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Guillermo Juárez-Vega
- Red de Apoyo a la Investigación, Coordinacion de Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandro Culebro-Bermejo
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| | - Diana Gómez-Martín
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán
| |
Collapse
|
6
|
Low-dose decitabine modulates myeloid-derived suppressor cell fitness via LKB1 in immune thrombocytopenia. Blood 2022; 140:2818-2834. [PMID: 36037415 DOI: 10.1182/blood.2022016029] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 08/11/2022] [Accepted: 08/11/2022] [Indexed: 01/05/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are heterogeneous immature cells and natural inhibitors of adaptive immunity. Metabolic fitness of MDSCs is fundamental for its suppressive activity toward effector T cells. Our previous studies showed that the number and inhibitory function of MDSCs were impaired in patients with immune thrombocytopenia (ITP) compared with healthy controls. In this study, we analyzed the effects of decitabine on MDSCs from patients with ITP, both in vitro and in vivo. We found that low-dose decitabine promoted the generation of MDSCs and enhanced their aerobic metabolism and immunosuppressive functions. Lower expression of liver kinase 1 (LKB1) was found in MDSCs from patients with ITP, which was corrected by decitabine therapy. LKB1 short hairpin RNA (shRNA) transfection effectively blocked the function of MDSCs and almost offset the enhanced effect of decitabine on impaired MDSCs. Subsequently, anti-CD61 immune-sensitized splenocytes were transferred into severe combined immunodeficient (SCID) mice to induce ITP in murine models. Passive transfer of decitabine-modulated MDSCs significantly raised platelet counts compared with that of phosphate buffered saline-modulated MDSCs. However, when LKB1 shRNA-transfected MDSCs were transferred into SCID mice, the therapeutic effect of decitabine in alleviating thrombocytopenia was quenched. In conclusion, our study suggests that the impaired aerobic metabolism of MDSCs is involved in the pathogenesis of ITP, and the modulatory effect of decitabine on MDSC metabolism contributes to the improvement of its immunosuppressive function. This provides a possible mechanism for sustained remission elicited by low-dose decitabine in patients with ITP.
Collapse
|
7
|
Tan L, Shi G, Zhao J, Xia X, Li D, Wang S, Liang J, Hou Y, Dou H. MDSCs participate in the pathogenesis of diffuse pulmonary hemorrhage in murine lupus through mTOR-FoxO1 signaling. Biochem Biophys Rep 2022; 32:101351. [PMID: 36164563 PMCID: PMC9507990 DOI: 10.1016/j.bbrep.2022.101351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Liping Tan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Guoping Shi
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Junyu Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Xiaoyu Xia
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Dan Li
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Saiwen Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, PR China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
- Corresponding author. The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China.
| | - Huan Dou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China
- Jiangsu Key Laboratory of Molecular Medicine, Nanjing, 210093, PR China
- Corresponding author. The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, Nanjing, 210093, PR China.
| |
Collapse
|
8
|
Yao G, Qi J, Li X, Tang X, Li W, Chen W, Xia N, Wang S, Sun L. Mesenchymal stem cell transplantation alleviated atherosclerosis in systemic lupus erythematosus through reducing MDSCs. Stem Cell Res Ther 2022; 13:328. [PMID: 35850768 PMCID: PMC9290280 DOI: 10.1186/s13287-022-03002-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/19/2022] [Indexed: 12/02/2022] Open
Abstract
Objective The mechanism by which mesenchymal stem cell (MSC) transplantation alleviates atherosclerosis in systemic lupus erythematosus (SLE) remains elusive. In this study, we aim to explore the efficacy and mechanism of MSC in ameliorating atherosclerosis in SLE. Methods ApoE−/− and Fas−/− mice on the B6 background were cross-bred to generate SLE mice with atherosclerosis. Myeloid-derived suppressor cells (MDSCs) were sorted and quantified. The apoE−/−Fas−/− mice were either treated with anti-Gr antibody or injected with MDSCs. The lupus-like autoimmunity and atherosclerotic lesions were evaluated. Furthermore, the apoE−/−Fas−/− mice were transplanted with MSCs and lupus-like autoimmunity and atherosclerotic lesions were assessed. Results MDSCs in peripheral blood, spleen, draining lymph nodes increased in apoE−/−Fas−/− mice compared with B6 mice. Moreover, the adoptive transfer of MDSCs aggravated both atherosclerosis and SLE pathologies, whereas depleting MDSCs ameliorated those pathologies in apoE−/−Fas−/− mice. MSC transplantation in apoE−/−Fas−/− mice decreased the percentage of MDSCs, alleviated the typical atherosclerotic lesions, including atherosclerotic lesions in aortae and liver, and reduced serum cholesterol, triglyceride and low-density lipoprotein levels. MSC transplantation also reduced SLE pathologies, including splenomegaly, glomerular lesions, anti-dsDNA antibody in serum, urine protein and serum creatinine. Moreover, MSC transplantation regulated the generation and function of MDSCs through secreting prostaglandin E 2 (PGE2). Conclusion Taken together, these results indicated that the increased MDSCs contributed to atherosclerosis in SLE. MSC transplantation ameliorated the atherosclerosis and SLE through reducing MDSCs by secreting PGE2. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03002-y.
Collapse
Affiliation(s)
- Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jingjing Qi
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.,Department of Immunology, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiaojing Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Wenchao Li
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Weiwei Chen
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Nan Xia
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Shiying Wang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| |
Collapse
|
9
|
Hou Y, Xie J, Wang S, Li D, Wang L, Wang H, Ni X, Leng S, Li G, Hou M, Peng J. Glucocorticoid receptor modulates myeloid-derived suppressor cell function via mitochondrial metabolism in immune thrombocytopenia. Cell Mol Immunol 2022; 19:764-776. [PMID: 35414712 PMCID: PMC9243139 DOI: 10.1038/s41423-022-00859-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature cells and natural inhibitors of adaptive immunity. Intracellular metabolic changes in MDSCs exert a direct immunological influence on their suppressive activity. Our previous study demonstrated that high-dose dexamethasone (HD-DXM) corrected the functional impairment of MDSCs in immune thrombocytopenia (ITP); however, the MDSC population was not restored in nonresponders, and the mechanism remained unclear. In this study, altered mitochondrial physiology and reduced mitochondrial gene transcription were detected in MDSCs from HD-DXM nonresponders, accompanied by decreased levels of carnitine palmitoyltransferase-1 (CPT-1), a rate-limiting enzyme in fatty acid oxidation (FAO). Blockade of FAO with a CPT-1 inhibitor abolished the immunosuppressive function of MDSCs in HD-DXM responders. We also report that MDSCs from ITP patients had lower expression of the glucocorticoid receptor (GR), which can translocate into mitochondria to regulate the transcription of mitochondrial DNA (mtDNA) as well as the level of oxidative phosphorylation. It was confirmed that the expression of CPT-1 and mtDNA-encoded genes was downregulated in GR-siRNA-treated murine MDSCs. Finally, by establishing murine models of active and passive ITP via adoptive transfer of DXM-modulated MDSCs, we confirmed that GR-silenced MDSCs failed to alleviate thrombocytopenia in mice with ITP. In conclusion, our study indicated that impaired aerobic metabolism in MDSCs participates in the pathogenesis of glucocorticoid resistance in ITP and that intact control of MDSC metabolism by GR contributes to the homeostatic regulation of immunosuppressive cell function.
Collapse
Affiliation(s)
- Yu Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Jie Xie
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuwen Wang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Daqi Li
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lingjun Wang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haoyi Wang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaofei Ni
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shaoqiu Leng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guosheng Li
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
- Advanced Medical Research Institute, Shandong University, Jinan, China.
| |
Collapse
|
10
|
Wang Y, Yan C, Su C, Wang Y, Luo S, Lu J, Zhao C, Zhao G, Xi J. Increased Frequency of Myeloid-Derived Suppressor Cells in Myasthenia Gravis After Immunotherapy. Front Neurol 2022; 13:902384. [PMID: 35847216 PMCID: PMC9278661 DOI: 10.3389/fneur.2022.902384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a population of myeloid progenitor cells with immunoregulatory functions and their role in myasthenia gravis (MG) was unknown. In this study, we investigated the phenotypic and functional alterations of MDSCs in MG before and after immunotherapy. The frequency of MDSCs significantly increased and negatively correlated to that of Th1 or Th17 cells after immunotherapy. MDSCs from untreated patients with MG showed an impaired suppression of IFN-γ production in T-cells and improved immunosuppressive function was identified after immunotherapy. The MFI of Arg-1 in MDSCs also increased after immunotherapy. These findings suggested the functional difference in MDSCs before and after immunotherapy, and MDSCs might play a role in disease remission.
Collapse
Affiliation(s)
- Yan Wang
- Central Lab, Huashan Hospital, Fudan University, Shanghai, China
| | - Chong Yan
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai, China
| | - Caixia Su
- China Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ying Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Sushan Luo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai, China
| | - Jun Lu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai, China
| | - Gan Zhao
- China Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Gan Zhao
| | - Jianying Xi
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Huashan Rare Disease Center, Shanghai, China
- Jianying Xi
| |
Collapse
|
11
|
Kustermann M, Dasari P, Knape I, Keltsch E, Liu J, Pflüger S, Osen W, Holzmann K, Huber-Lang M, Debatin KM, Strauss G. Adoptively Transferred in vitro-Generated Myeloid-Derived Suppressor Cells Improve T-Cell Function and Antigen-Specific Immunity after Traumatic Lung Injury. J Innate Immun 2022; 15:78-95. [PMID: 35691281 PMCID: PMC10643914 DOI: 10.1159/000525088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/07/2022] [Indexed: 11/19/2022] Open
Abstract
Immune reactions after trauma are characterized by immediate activation of innate immunity and simultaneously downregulation of adaptive immunity leading to a misbalanced immunohomeostasis and immunosuppression of the injured host. Therefore, the susceptibility to secondary infections is strongly increased after trauma. Immune responses are regulated by a network of immune cells influencing each other and at the same time modifying their functions dependent on the inflammatory environment. Although myeloid-derived suppressor cells (MDSCs) are initially described as T-cell suppressors, their immunomodulatory capacity after trauma is mostly undefined. Therefore, in vitro-generated MDSCs were adoptively transferred into mice after blunt chest trauma (TxT). A single MDSC treatment-induced splenic T-cell expansion decreased apoptosis sensitivity and improved proliferation in the absence of T-cell exhaustion until 2 weeks after trauma. MDSC treatment had a long-lasting effect on the genomic landscape of CD4+ T cells by upregulating primarily Th2-associated genes. Remarkably, immune-activating functions of MDSCs supported the ability of TxT mice to respond to post-traumatic secondary antigen challenge. Secondary insults were mimicked by immunizing MDSC-treated TxT mice with ovalbumin (OVA), followed by OVA restimulation in vitro. MDSC treatment significantly increased the frequency of OVA-specific T cells, enhanced their Th1/Th2 cytokine expression, and induced upregulation of cytolytic molecules finally improving OVA-specific cytotoxicity. Overall, we could show that therapeutic MDSC treatment after TxT improves post-traumatic T-cell functions, which might enable the traumatic host to counterbalance trauma-induced immunoparalysis.
Collapse
Affiliation(s)
- Monika Kustermann
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Prasad Dasari
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Ingrid Knape
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Emma Keltsch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Jianing Liu
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Silvia Pflüger
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Wolfram Osen
- GMP & T Cell Therapy, German Cancer Research Center, Heidelberg, Germany
| | | | - Markus Huber-Lang
- Institute of Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
12
|
Grassi G, Notari S, Gili S, Bordoni V, Casetti R, Cimini E, Tartaglia E, Mariotti D, Agrati C, Sacchi A. Myeloid-Derived Suppressor Cells in COVID-19: The Paradox of Good. Front Immunol 2022; 13:842949. [PMID: 35572540 PMCID: PMC9092297 DOI: 10.3389/fimmu.2022.842949] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/25/2022] [Indexed: 12/26/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing coronavirus disease 2019 (COVID-19) pandemic. Viral replication in the respiratory tract induces the death of infected cells and the release of pathogen- associated molecular patterns (PAMPs). PAMPs give rise to local inflammation, increasing the secretion of pro- inflammatory cytokines and chemokines, which attract immune cells from the blood into the infected lung. In most individuals, lung-recruited cells clear the infection, and the immune response retreats. However, in some cases, a dysfunctional immune response occurs, which triggers a cytokine storm in the lung, leading to acute respiratory distress syndrome (ARDS). Severe COVID-19 is characterized by an impaired innate and adaptive immune response and by a massive expansion of myeloid-derived suppressor cells (MDSCs). MDSCs function as protective regulators of the immune response, protecting the host from over-immunoreactivity and hyper-inflammation. However, under certain conditions, such as chronic inflammation and cancer, MDSCs could exert a detrimental role. Accordingly, the early expansion of MDSCs in COVID-19 is able to predict the fatal outcome of the infection. Here, we review recent data on MDSCs during COVID-19, discussing how they can influence the course of the disease and whether they could be considered as biomarker and possible targets for new therapeutic approaches.
Collapse
Affiliation(s)
- Germana Grassi
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Stefania Notari
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Simona Gili
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Veronica Bordoni
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Rita Casetti
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Eleonora Cimini
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Eleonora Tartaglia
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Davide Mariotti
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Chiara Agrati
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Alessandra Sacchi
- Laboratory of Cellular Immunology and Pharmacology, National Institute for infectious Diseases "Lazzaro Spallanzani"-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| |
Collapse
|
13
|
Horwitz JK, Bin S, Fairchild RL, Keslar KS, Yi Z, Zhang W, Pavlov VI, Li Y, Madsen JC, Cravedi P, Heeger PS. Linking erythropoietin to regulatory T-cell-dependent allograft survival through myeloid cells. JCI Insight 2022; 7:158856. [PMID: 35389892 PMCID: PMC9220923 DOI: 10.1172/jci.insight.158856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/06/2022] [Indexed: 12/03/2022] Open
Abstract
Erythropoietin (EPO) has multiple nonerythropoietic functions, including immune modulation, but EPO’s effects in transplantation remain incompletely understood. We tested the mechanisms linking EPO administration to prolongation of murine heterotopic heart transplantation using WT and conditional EPO receptor–knockout (EPOR-knockout) mice as recipients. In WT controls, peritransplant administration of EPO synergized with CTLA4-Ig to prolong allograft survival (P < 0.001), reduce frequencies of donor-reactive effector CD8+ T cells in the spleen (P < 0.001) and in the graft (P < 0.05), and increase frequencies and total numbers of donor-reactive Tregs (P < 0.01 for each) versus CTLA4-Ig alone. Studies performed in conditional EPOR-knockout recipients showed that each of these differences required EPOR expression in myeloid cells but not in T cells. Analysis of mRNA isolated from spleen monocytes showed that EPO/EPOR ligation upregulated macrophage-expressed, antiinflammatory, regulatory, and pro-efferocytosis genes and downregulated selected proinflammatory genes. Taken together, the data support the conclusion that EPO promotes Treg-dependent murine cardiac allograft survival by crucially altering the phenotype and function of macrophages. Coupled with our previous documentation that EPO promotes Treg expansion in humans, the data support the need for testing the addition of EPO to costimulatory blockade-containing immunosuppression regimens in an effort to prolong human transplant survival.
Collapse
Affiliation(s)
- Julian K Horwitz
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Sofia Bin
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Robert L Fairchild
- Department of Immunology, Cleveland Clinic, Cleveland, United States of America
| | - Karen S Keslar
- Department of Immunology, Cleveland Clinic, Cleveland, United States of America
| | - Zhengzi Yi
- Translational Transplant Research Center, Icahn School of medicine at Mount Sinai, New York, United States of America
| | - Weijia Zhang
- Translational Transplant Research Center, Icahn school of Medicine at Mount Sinai, New York, United States of America
| | - Vasile I Pavlov
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Yansui Li
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Joren C Madsen
- Department of Surgery, Massachusetts General Hospital, Boston, United States of America
| | - Paolo Cravedi
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| | - Peter S Heeger
- Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, New York, United States of America
| |
Collapse
|
14
|
Boral B, Ballı HT, Sözütok S, Pehlivan UA, Aikimbaev K. Clinical and prognostic significance of CD14 (+) HLA-DR (-/low) myeloid-derived suppressor cells in patients with hepatocellular carcinoma received transarterial radioembolization with Yttrium-90. Scand J Immunol 2021; 95:e13132. [PMID: 34936119 DOI: 10.1111/sji.13132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 11/21/2021] [Accepted: 12/15/2021] [Indexed: 01/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide. For unresectable HCC, transarterial radioembolization (TARE) with Yttrium-90 is a widely used treatment. The aim of this study was to investigate whether monocytic myeloid-derived suppressor cells (M-MDSC) and CD39+ T cells can be non-invasive predictive biomarkers of radiological response and prognosis in patients with HCC treated with TARE. This study was conducted on 39 patients with HCC who were treated with TARE between August 2018 and December 2019 and the control group consisted of 23 healthy volunteers. CD4+, CD8+, CD39+ T cells, Natural killer (NK) cells, myeloid cells (MC) and M-MDSC parameters are examined in the course of TARE treatment with student t test and Kaplan-Meier method. There were statistically significant differences in M-MDSC, CD39+ T cells and MC values between healthy controls and HCC patients. A statistically significant difference was found in M-MDSC and CD4+ T cells values in the HCC patient group who responded to the treatment compared to those who did not. Survival analysis found that patients with lower frequencies (under 3.81%) of M-MDSC showed more prominent differences of overall survival (OS) compared to patients with all high groups. We found that M-MDSC in the peripheral blood might be a useful non-invasive biomarker to predict OS. We have shown for the first time that M-MDSC is correlated with treatment response in HCC patients treated with TARE. Additionally, we have found that the percentage of CD39+ T cells is high in HCC patients and these cells are positively correlated with M-MDSC.
Collapse
Affiliation(s)
- Barış Boral
- Department of Immunology, Adana Health Practice and Research Center, University of Health Sciences, Adana, Turkey
| | | | - Sinan Sözütok
- Department of Radiology, Çukurova University School of Medicine, Adana, Turkey
| | - Umur Anıl Pehlivan
- Department of Radiology, Çukurova University School of Medicine, Adana, Turkey
| | - Kairgeldy Aikimbaev
- Department of Radiology, Çukurova University School of Medicine, Adana, Turkey
| |
Collapse
|
15
|
Bareke H, Juanes-Velasco P, Landeira-Viñuela A, Hernandez AP, Cruz JJ, Bellido L, Fonseca E, Niebla-Cárdenas A, Montalvillo E, Góngora R, Fuentes M. Autoimmune Responses in Oncology: Causes and Significance. Int J Mol Sci 2021; 22:ijms22158030. [PMID: 34360795 PMCID: PMC8347170 DOI: 10.3390/ijms22158030] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/19/2021] [Accepted: 07/23/2021] [Indexed: 12/20/2022] Open
Abstract
Specific anti-tumor immune responses have proven to be pivotal in shaping tumorigenesis and tumor progression in solid cancers. These responses can also be of an autoimmune nature, and autoantibodies can sometimes be present even before the onset of clinically overt disease. Autoantibodies can be generated due to mutated gene products, aberrant expression and post-transcriptional modification of proteins, a pro-immunogenic milieu, anti-cancer treatments, cross-reactivity of tumor-specific lymphocytes, epitope spreading, and microbiota-related and genetic factors. Understanding these responses has implications for both basic and clinical immunology. Autoantibodies in solid cancers can be used for early detection of cancer as well as for biomarkers of prognosis and treatment response. High-throughput techniques such as protein microarrays make parallel detection of multiple autoantibodies for increased specificity and sensitivity feasible, affordable, and quick. Cancer immunotherapy has revolutionized cancer treatments and has made a considerable impact on reducing cancer-associated morbidity and mortality. However, immunotherapeutic interventions such as immune checkpoint inhibition can induce immune-related toxicities, which can even be life-threatening. Uncovering the reasons for treatment-induced autoimmunity can lead to fine-tuning of cancer immunotherapy approaches to evade toxic events while inducing an effective anti-tumor immune response.
Collapse
Affiliation(s)
- Halin Bareke
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Institute of Health Sciences, Marmara University, Istanbul 34722, Turkey;
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Alicia Landeira-Viñuela
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Angela-Patricia Hernandez
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Juan Jesús Cruz
- Medical Oncology Service, Hospital Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain; (J.J.C.); (L.B.); (E.F.)
| | - Lorena Bellido
- Medical Oncology Service, Hospital Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain; (J.J.C.); (L.B.); (E.F.)
| | - Emilio Fonseca
- Medical Oncology Service, Hospital Universitario de Salamanca-IBSAL, 37007 Salamanca, Spain; (J.J.C.); (L.B.); (E.F.)
| | - Alfonssina Niebla-Cárdenas
- Department of Nursing and Physiotherapy, Faculty of Nursing and Physiotherapy, University of Salamanca, 37007 Salamanca, Spain;
| | - Enrique Montalvillo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Rafael Góngora
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain; (P.J.-V.); (A.L.-V.); (A.-P.H.); (E.M.); (R.G.)
- Proteomics Unit, Cancer Research Center (IBMCC/CSIC/USAL/IBSAL), 37007 Salamanca, Spain
- Correspondence: ; Tel.: +34-923-294-811
| |
Collapse
|
16
|
Role of Myeloid-derived suppressor cell (MDSC) in autoimmunity and its potential as a therapeutic target. Inflammopharmacology 2021; 29:1307-1315. [PMID: 34283371 DOI: 10.1007/s10787-021-00846-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/04/2021] [Indexed: 02/07/2023]
Abstract
Myeloid suppressor cells (MDSCs) are an important class of immune-regulating cells that can suppress T cell function. Most of our knowledge about the function of MDSC comes from studies of cancer models. Recent studies, however, have greatly contributed to the description of MDSC involvement in autoimmune diseases. They are known as a cell population that may negatively affect immune responses by regulating the function of CD4+ and CD8+ cells, which makes them an attractive target for autoimmune diseases therapy. However, many questions about MDSC activation, differentiation, and inhibitory functions remain unanswered. In this study, we have summarized the role of MDSCs in various autoimmune diseases, and the potential of targeting them for therapeutic benefits has been discussed.
Collapse
|
17
|
Dubik M, Marczynska J, Mørch MT, Webster G, Jensen KN, Wlodarczyk A, Khorooshi R, Owens T. Innate Signaling in the CNS Prevents Demyelination in a Focal EAE Model. Front Neurosci 2021; 15:682451. [PMID: 34149350 PMCID: PMC8209300 DOI: 10.3389/fnins.2021.682451] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/07/2021] [Indexed: 11/13/2022] Open
Abstract
The pathological hallmark of multiple sclerosis (MS) is the formation of multifocal demyelinating lesions in the central nervous system (CNS). Stimulation of innate receptors has been shown to suppress experimental autoimmune encephalomyelitis (EAE), an MS-like disease in mice. Specifically, targeting Toll-like receptor 9 (TLR9) and NOD-like receptor 2 (NOD2) significantly reduced disease severity. In the present work we have developed a novel focal EAE model to further study the effect of innate signaling on demyelinating pathology. Focal lesions were induced by stereotactic needle insertion into the corpus callosum (CC) of mice previously immunized for EAE. This resulted in focal pathology characterized by infiltration and demyelination in the CC. We find that intrathecal delivery of MIS416, a TLR9 and NOD2 bispecific innate ligand, into the cerebrospinal fluid reduced focal lesions in the CC. This was associated with upregulation of type I and II interferons, interleukin-10, arginase-1, CCL-2 and CXCL-10. Analysis of draining cervical lymph nodes showed upregulation of type II interferons and interleukin 10. Moreover, intrathecal MIS416 altered the composition of early CNS infiltrates, increasing proportions of myeloid and NK cells and reducing T cells at the lesion site. This study contributes to an increased understanding of how innate immune responses can play a protective role, which in turn may lead to additional therapeutic strategies for the prevention and treatment of demyelinating pathologies.
Collapse
Affiliation(s)
- Magdalena Dubik
- Neurobiology, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Joanna Marczynska
- Neurobiology, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Marlene T Mørch
- Neurobiology, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Gill Webster
- Innate Immunotherapeutics, Auckland, New Zealand
| | - Kirstine Nolling Jensen
- Neurobiology, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Agnieszka Wlodarczyk
- Neurobiology, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Reza Khorooshi
- Neurobiology, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Trevor Owens
- Neurobiology, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
18
|
Role of Regulatory Cells in Immune Tolerance Induction in Hemophilia A. Hemasphere 2021; 5:e557. [PMID: 33898928 PMCID: PMC8061682 DOI: 10.1097/hs9.0000000000000557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/09/2021] [Indexed: 11/26/2022] Open
Abstract
The main complication of hemophilia A treatment is the development of neutralizing antibodies (inhibitors) against factor VIII (FVIII). Immune tolerance induction (ITI) is the prescribed treatment for inhibitor eradication, although its working mechanism remains unresolved. To clarify this mechanism, we compared blood samples of hemophilia A patients with and without inhibitors for presence of immunoregulatory cells and markers, including regulatory B-cells (Bregs), regulatory T-cells (Tregs), myeloid-derived suppressor cells (MDSCs), and expression of regulatory markers on T-cells (programmed cell death protein 1 [PD1], inducable T-cell costimulator, cytotoxic T-lymphocyte-associated protein 4 [CTLA4]), by use of flow cytometry. By cross-sectional analysis inhibitor patients (N = 20) were compared with inhibitor-negative (N = 28) and ex-inhibitor (N = 17) patients. In another longitudinal study, changes in immunoregulatory parameters were evaluated during ITI (N = 12) and compared with inhibitor-negative hemophilia A patients (N = 36). The frequency of Bregs, but not of Tregs nor MDSCs, was significantly reduced in inhibitor patients (3.2%) compared with inhibitor-negative (5.9%) and ex-inhibitor patients (8.9%; P < 0.01). CTLA4 expression on T-cells was also reduced (mean fluorescence intensity 133 in inhibitor versus 537 in inhibitor-negative patients; P < 0.01). Fittingly, in patients followed during ITI, inhibitor eradication associated with increased Bregs, increased Tregs, and increased expression of CTLA4 and PD1 on CD4+ T-cells. In conclusion, inhibitor patients express significantly lower frequency of Bregs and Tregs marker expression, which are restored by successful ITI. Our findings suggest that an existing anti-FVIII immune response is associated with deficits in peripheral tolerance mechanisms and that Bregs and changes in immunoregulatory properties of CD4+ T-cells likely contribute to ITI in hemophilia A patients with inhibitors.
Collapse
|
19
|
MacDonald C, Ministero S, Pandey M, Robinson D, Forti Hong E, Hylander B, McCarthy P, Gordon C, Repasky E, Mohammadpour H. Comparing thermal stress reduction strategies that influence MDSC accumulation in tumor bearing mice. Cell Immunol 2021; 361:104285. [PMID: 33484943 PMCID: PMC7883813 DOI: 10.1016/j.cellimm.2021.104285] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022]
Abstract
Myeloid derived suppressor cells (MDSCs) are a diverse collection of immune cells that suppress anti-tumor immune responses. Decreasing MDSCs accumulation in the tumor microenvironment could improve the anti-tumor immune response and improve immunotherapy. Here, we examine the impact of physiologically relevant thermal treatments on the accumulation of MDSCs in tumors in mice. We found that different temperature-based protocols, including 1) weekly whole-body hyperthermia, 2) housing mice at their thermoneutral temperature (TT, ~30 °C), and 3) housing mice at a subthermoneutral temperature (ST,~22 °C) while providing a localized heat source, each resulted in a reduction in MDSC accumulation and improved tumor growth control compared to control mice housed at ST, which is the standard, mandated housing temperature for laboratory mice. Additionally, we found that low dose β-adrenergic receptor blocker (propranolol) therapy reduced MDSC accumulation and improved tumor growth control to a similar degree as the models that relieved cold stress. These results show that thermal treatments can decrease MDSC accumulation and tumor growth comparable to propranolol therapy.
Collapse
Affiliation(s)
- Cameron MacDonald
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Samuel Ministero
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Manu Pandey
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Denisha Robinson
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Evan Forti Hong
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Bonnie Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Philip McCarthy
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | | | - Elizabeth Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States.
| | - Hemn Mohammadpour
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States.
| |
Collapse
|
20
|
Rambault M, Doz-Deblauwe É, Le Vern Y, Carreras F, Cunha P, Germon P, Rainard P, Winter N, Remot A. Neutrophils Encompass a Regulatory Subset Suppressing T Cells in Apparently Healthy Cattle and Mice. Front Immunol 2021; 12:625244. [PMID: 33717136 PMCID: PMC7952614 DOI: 10.3389/fimmu.2021.625244] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
Abstract
Neutrophils that reside in the bone marrow are swiftly recruited from circulating blood to fight infections. For a long time, these first line defenders were considered as microbe killers. However their role is far more complex as cross talk with T cells or dendritic cells have been described for human or mouse neutrophils. In cattle, these new roles are not documented yet. We identified a new subset of regulatory neutrophils that is present in the mouse bone marrow or circulate in cattle blood under steady state conditions. These regulatory neutrophils that display MHC-II on the surface are morphologically indistinguishable from classical MHC-IIneg neutrophils. However MHC-IIpos and MHC-IIneg neutrophils display distinct transcriptomic profiles. While MHC-IIneg and MHC-IIpos neutrophils display similar bacterial phagocytosis or killing activity, MHC-IIpos only are able to suppress T cell proliferation under contact-dependent mechanisms. Regulatory neutrophils are highly enriched in lymphoid organs as compared to their MHC-IIneg counterparts and in the mouse they express PDL-1, an immune checkpoint involved in T-cell blockade. Our results emphasize neutrophils as true partners of the adaptive immune response, including in domestic species. They open the way for discovery of new biomarkers and therapeutic interventions to better control cattle diseases.
Collapse
Affiliation(s)
- Marion Rambault
- INRAE, Université de Tours, ISP, Nouzilly, France.,Institut de l'Elevage, Paris, France
| | | | - Yves Le Vern
- INRAE, Université de Tours, ISP, Nouzilly, France
| | | | | | | | | | | | - Aude Remot
- INRAE, Université de Tours, ISP, Nouzilly, France
| |
Collapse
|
21
|
Blanter M, Gouwy M, Struyf S. Studying Neutrophil Function in vitro: Cell Models and Environmental Factors. J Inflamm Res 2021; 14:141-162. [PMID: 33505167 PMCID: PMC7829132 DOI: 10.2147/jir.s284941] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/04/2020] [Indexed: 01/21/2023] Open
Abstract
Neutrophils are the most abundant immune cell type in the blood and constitute the first line of defense against invading pathogens. Despite their important role in many diseases, they are challenging to study due to their short life span and the inability to cryopreserve or expand them in vitro. Thus, research into neutrophils has to rely on cells freshly isolated from peripheral blood of human donors, introducing donor-dependent variation in the experimental data. To counteract these problems, researchers tried to develop adequate cell models, such as cell lines. For those functional studies that cannot rely on cell models, a standardization of protocols regarding neutrophil purification and culturing could be a solution. In this review, we provide an overview of the most commonly used models for neutrophil function (HL-60, PLB-985, NB4, Kasumi-1 and induced pluripotent stem cells). In addition, we describe the effects of glucose concentration, pH, oxygen tension and temperature on neutrophil function.
Collapse
Affiliation(s)
- Marfa Blanter
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium
| | - Mieke Gouwy
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, University of Leuven, Leuven 3000, Belgium
| |
Collapse
|
22
|
Michaud D, Steward CR, Mirlekar B, Pylayeva-Gupta Y. Regulatory B cells in cancer. Immunol Rev 2021; 299:74-92. [PMID: 33368346 PMCID: PMC7965344 DOI: 10.1111/imr.12939] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/25/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022]
Abstract
Tumorigenesis proceeds through discrete steps where acquisition of genetic lesions and changes in the surrounding microenvironment combine to drive unrestricted neoplastic proliferation and metastasis. The ability of tumor-infiltrating immune cells to promote tumor growth via the provision of signals that enable tumor cell survival and proliferation as well as contribute to immune suppression is an active area of research. Recent efforts have provided us with mechanistic insights into how B cells can positively and negatively regulate immune responses. Negative regulation of immune responses in cancer can be mediated by regulatory B cells and is often a result of increased production of cytokines that can directly and indirectly affect anti-tumor immune function and cancer cell growth. Signals that lead to the expansion of regulatory B cells and the spectrum of their functional roles are not well understood and are the subject of active research by many groups. Here, we elaborate broadly on the history of regulatory B cells in cancer and summarize recent studies that have established genetic models for the study of regulatory B cell function and their potential for therapeutic intervention in the setting of solid cancers.
Collapse
Affiliation(s)
- Daniel Michaud
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Colleen R Steward
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Bhalchandra Mirlekar
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Genetics, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Yuliya Pylayeva-Gupta
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Genetics, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
23
|
Wu Y, Luo J, Garden OA. Immunoregulatory Cells in Myasthenia Gravis. Front Neurol 2020; 11:593431. [PMID: 33384654 PMCID: PMC7769807 DOI: 10.3389/fneur.2020.593431] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/23/2020] [Indexed: 12/22/2022] Open
Abstract
Myasthenia gravis (MG) is a T cell-dependent, B-cell mediated autoimmune disease caused by antibodies against the nicotinic acetylcholine receptor or other components of the post-synaptic muscle endplate at the neuromuscular junction. These specific antibodies serve as excellent biomarkers for diagnosis, but do not adequately substitute for clinical evaluations to predict disease severity or treatment response. Several immunoregulatory cell populations are implicated in the pathogenesis of MG. The immunophenotype of these populations has been well-characterized in human peripheral blood. CD4+FoxP3+ regulatory T cells (Tregs) are functionally defective in MG, but there is a lack of consensus on whether they show numerical perturbations. Myeloid-derived suppressor cells (MDSCs) have also been explored in the context of MG. Adoptive transfer of CD4+FoxP3+ Tregs or MDSCs suppresses ongoing experimental autoimmune MG (EAMG), a rodent model of MG, suggesting a protective role of both populations in this disease. An imbalance between follicular Tregs and follicular T helper cells is found in untreated MG patients, correlating with disease manifestations. There is an inverse correlation between the frequency of circulating IL-10–producing B cells and clinical status in MG patients. Taken together, both functional and numerical defects in various populations of immunoregulatory cells in EAMG and human MG have been demonstrated, but how they relate to pathogenesis and whether these cells can serve as biomarkers of disease activity in humans deserve further exploration.
Collapse
Affiliation(s)
- Ying Wu
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jie Luo
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Oliver A Garden
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
24
|
The importance of advanced cytometry in defining new immune cell types and functions relevant for the immunopathogenesis of HIV infection. AIDS 2020; 34:2169-2185. [PMID: 32910071 DOI: 10.1097/qad.0000000000002675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
: In the last years, novel, exciting immunological findings of interest for HIV research and treatment were identified thanks to different cytometric approaches. The analysis of the phenotypes and functionality of cells belonging to the immune system could clarify their role in the immunopathogenesis of HIV infection, and to elaborate key concepts, relevant in the treatment of this disease. Important discoveries have been made concerning cells that are important for protective immunity like lymphocytes that display polyfunctionality, resident memory T cells, innate lymphoid cells, to mention a few. The complex phenotype of myeloid-derived suppressor cells has been investigated, and relevant changes have been reported during chronic and primary HIV infection, in correlation with changes in CD4 T-cell number, T-cell activation, and with advanced disease stage. The search for markers of HIV persistence present in latently infected cells, namely those molecules that are important for a functional or sterilizing cure, evidenced the role of follicular helper T cells, and opened a discussion on the meaning and use of different surface molecules not only in identifying such cells, but also in designing new strategies. Finally, advanced technologies based upon the simultaneous detection of HIV-RNA and proteins at the single cell level, as well as those based upon spectral cytometry or mass cytometry are now finding new actors and depicting a new scenario in the immunopathogenesis of the infection, that will allow to better design innovative therapies based upon novel drugs and vaccines.
Collapse
|
25
|
Wu Y, Chang YM, Lawson BS, Galban EM, Mittelman NS, Benedicenti L, Petesch SC, Carroll AB, Punt JA, Luo J, Garden OA. Myeloid-derived suppressor cell and regulatory T cell frequencies in canine myasthenia gravis: A pilot study. Vet J 2020; 267:105581. [PMID: 33375962 DOI: 10.1016/j.tvjl.2020.105581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 11/15/2020] [Accepted: 11/23/2020] [Indexed: 01/21/2023]
Abstract
Myasthenia gravis (MG) is a T cell-dependent, B cell-mediated autoimmune disease. Little is known about its cellular pathogenesis in dogs. This study provides the first preliminary assessment of the frequency of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) in the peripheral blood of dogs with seropositive generalized MG. No alteration in frequency of either MDSCs or Tregs in dogs with MG was observed when compared to those in either seronegative dogs with diagnoses other than MG, or healthy dogs. A longitudinal study in three dogs with MG revealed no correlation between the relative numbers of either population and the clinical course of disease. Neither the frequency of MDSCs nor of Tregs showed a correlation with anti-AChR antibody titer in dogs with MG. These findings suggest that aberrations in the frequency of either immunosuppressive population do not occur in MG, but they need to be validated in large-scale prospective studies.
Collapse
Affiliation(s)
- Ying Wu
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Yu-Mei Chang
- Research Support Office, Royal Veterinary College, London NW1 0TU, UK
| | - Brandon S Lawson
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Evelyn M Galban
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Neil S Mittelman
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Leontine Benedicenti
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Scott C Petesch
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Alicia B Carroll
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jennifer A Punt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Jie Luo
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | - Oliver A Garden
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
26
|
Ramachandran P, Kathirvelu B, Chakraborti A, Gajendran M, Zhahid U, Ghanta S, Onukogu I, Narh JT, Wang JC, Anwer F. COVID-19 in Cancer Patients From New York City: A Comparative Single Center Retrospective Analysis. Cancer Control 2020; 27:1073274820960457. [PMID: 33043705 PMCID: PMC7791463 DOI: 10.1177/1073274820960457] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In this retrospective study we analyze and compare clinical characteristics and
outcomes of patients with and without cancer history who were infected with
novel coronavirus disease 19 (COVID-19). Medical records were reviewed and a
comparative analysis of 53 cancer and 135 non-cancer patients with COVID-19 were
summarized. Results: The median age for COVID-19 patients with and without
cancer was 71.5 and 61.6 years, respectively. Patients aged 60 years and above
were 86.8% and 60.7% in cancer and non-cancer groups, respectively. A high
proportion of cases were seen in African Americans 73.6% (with cancer) and 75.6%
(without cancer) followed by Hispanic patients. Male and female patients had a
high percentage of prostate (39.3%) and breast (32%) cancer respectively.
Prostate cancer (18.9%) and myeloma (11.3%) were common among solid and
hematological cancers respectively. Hypertension and smoking were prevalent
among cancer (83% and 41.5%) compared to non-cancer (67.4% and 9.6%) patients.
The common symptoms in cancer patients were dyspnea (64.2%) followed by fever
and cough (50.9%) compared to fever (68.1%) and cough (66.7%) in non-cancer
patients. Cancer patients had higher levels of lactic acidosis, C-reactive
protein, lactate dehydrogenase, and alkaline phosphatase than non-cancer
patients (p < 0.05). Conclusions: Rapid clinical deterioration was seen in
cancer patients who were aged 60 years and above. Higher mortality was seen in
this subgroup, especially when they had associated hypertension and elevated
levels of CRP and LDH.
Collapse
Affiliation(s)
- Preethi Ramachandran
- Department of Hematology and Oncology, 2025Brookdale University Hospital and Medical Center, Brooklyn, NY, USA
| | | | - Abhishek Chakraborti
- Department of Internal Medicine, 2025Brookdale University Hospital and Medical Center, Brooklyn, NY, USA
| | - Mahesh Gajendran
- Department of Internal Medicine, Paul L. Foster School of Medicine, 37316Texas Tech University, El Paso, TX, USA
| | - Umar Zhahid
- Department of Nephrology, 2025Brookdale University Hospital and Medical Center, Brooklyn, NY, USA
| | - Snigdha Ghanta
- Department of Internal Medicine, 2025Brookdale University Hospital and Medical Center, Brooklyn, NY, USA
| | - Ifeanyichkwu Onukogu
- Department of Internal Medicine, 2025Brookdale University Hospital and Medical Center, Brooklyn, NY, USA
| | - Joshua Tetteh Narh
- Department of Internal Medicine, 2025Brookdale University Hospital and Medical Center, Brooklyn, NY, USA
| | - Jen C Wang
- New York Medical College, Hematology/Oncology research laboratory, 2025Brookdale University Hospital and Medical Center, Brooklyn, NY, USA
| | - Faiz Anwer
- Tausig Cancer Center, Hematology/Oncology, Stem Cell Transplantation, 2569Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
27
|
Zhong L, Li S, Wen Y, Zheng J, Liu F, Cao D, Liu Y. Expansion of Polymorphonuclear Myeloid-Derived Suppressor Cells in Patients With Gout. Front Immunol 2020; 11:567783. [PMID: 33154749 PMCID: PMC7591715 DOI: 10.3389/fimmu.2020.567783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 09/10/2020] [Indexed: 12/31/2022] Open
Abstract
Gout is an inflammatory joint disease caused by monosodium urate (MSU) crystals; however, the mechanism underlying MSU-induced inflammation is unclear. Previous research has suggested that inflammation or cancer can drive the expansion of myeloid-derived suppressor cells (MDSCs). In this study, the role of MDSCs in MSU-induced gout inflammation was evaluated. A total of 28 patients with gout, and 20 healthy controls were recruited for the study. MDSCs, and their functions, were analyzed by flow cytometry and a T cell co-culture assay, respectively. We observed a higher frequency of PMN-MDSCs, and a stronger immunosuppressive function, in patients with gout compared to the controls. Moreover, circulating PMN-MDSCs were positively correlated with pathological indicators, including uric acid and C-reactive protein levels. We also demonstrated that MSU can induce significant PMN-MDSC expansion, using in vivo and in vitro experiments. Finally, MSU-induced PMN-MDSCs produced higher levels of IL-1β, which mediated gout inflammatory progression. Our results demonstrate that MSU modulates the expansion and suppressive function of PMN-MDSCs, providing insights into a novel mechanism underlying the pathogenesis of MSU-induced gout. Thus, MDSCs may be useful for the development of novel therapeutic strategies for the prevention and treatment of gout.
Collapse
Affiliation(s)
- Limei Zhong
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Sitao Li
- Department of Neonatology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi Wen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junhui Zheng
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengbin Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Donglin Cao
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Yufeng Liu
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangzhou First People's Hospital, Guangzhou, China
| |
Collapse
|
28
|
Owens T, Benmamar-Badel A, Wlodarczyk A, Marczynska J, Mørch MT, Dubik M, Arengoth DS, Asgari N, Webster G, Khorooshi R. Protective roles for myeloid cells in neuroinflammation. Scand J Immunol 2020; 92:e12963. [PMID: 32851668 DOI: 10.1111/sji.12963] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022]
Abstract
Myeloid cells represent the major cellular component of innate immune responses. Myeloid cells include monocytes and macrophages, granulocytes (neutrophils, basophils and eosinophils) and dendritic cells (DC). The role of myeloid cells has been broadly described both in physiological and in pathological conditions. All tissues or organs are equipped with resident myeloid cells, such as parenchymal microglia in the brain, which contribute to maintaining homeostasis. Moreover, in case of infection or tissue damage, other myeloid cells such as monocytes or granulocytes (especially neutrophils) can be recruited from the circulation, at first to promote inflammation and later to participate in repair and regeneration. This review aims to address the regulatory roles of myeloid cells in inflammatory diseases of the central nervous system (CNS), with a particular focus on recent work showing induction of suppressive function via stimulation of innate signalling in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE).
Collapse
Affiliation(s)
- Trevor Owens
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Anouk Benmamar-Badel
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark.,Department of Neurology, Slagelse Hospital, Slagelse, Denmark
| | - Agnieszka Wlodarczyk
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Joanna Marczynska
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Marlene T Mørch
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Magdalena Dubik
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Dina S Arengoth
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Nasrin Asgari
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark.,Department of Neurology, Slagelse Hospital, Slagelse, Denmark
| | - Gill Webster
- Innate Immunotherapeutics, Auckland, New Zealand
| | - Reza Khorooshi
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
29
|
Yan L, Liang M, Yang T, Ji J, Jose Kumar Sreena GS, Hou X, Cao M, Feng Z. The Immunoregulatory Role of Myeloid-Derived Suppressor Cells in the Pathogenesis of Rheumatoid Arthritis. Front Immunol 2020; 11:568362. [PMID: 33042149 PMCID: PMC7522347 DOI: 10.3389/fimmu.2020.568362] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/13/2020] [Indexed: 12/17/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of cells that regulate the immune response and exert immunosuppressive effects on various immune cells. Current studies indicate that MDSCs have both anti-inflammatory effects and proinflammatory effects on rheumatoid arthritis (RA) and RA animal models. MDSCs inhibit CD4+ T cells, which secrete proinflammatory factors such as IFN-γ, IL-2, IL-6, IL-17, and TNF-α, by inhibiting iNOS, ROS, and IFN-γ and promoting the production of the anti-inflammatory factor IL-10. MDSCs can suppress dendritic cells by reducing MHC-II and CD86 expression, expand Treg cells in vitro through the action of IL-10, inhibit B cells through NO and PGE2, and promote Th17 cell responses by secreting IL-1β. As a type of osteoclast precursor cell, MDSCs can differentiate into osteoclasts through activation of the NF-κB pathway via IL-1α. Overall, our study reviews the research progress related to MDSCs in RA, focusing on the effects of MDSCs on various types of cells and aiming to provide ideas to help reveal the important role of MDSCs in RA.
Collapse
Affiliation(s)
- Lan Yan
- Medical College of China Three Gorges University, Yichang, China
| | - Mingge Liang
- Medical College of China Three Gorges University, Yichang, China
| | - Tong Yang
- Medical College of China Three Gorges University, Yichang, China
| | - Jinyu Ji
- Medical College of China Three Gorges University, Yichang, China
| | | | - Xiaoqiang Hou
- The Institute of Rheumatology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
| | - Meiqun Cao
- Shenzhen Institute of Geriatrics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhitao Feng
- Medical College of China Three Gorges University, Yichang, China
- The Institute of Rheumatology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, China
| |
Collapse
|
30
|
Hager S, Fittler FJ, Wagner E, Bros M. Nucleic Acid-Based Approaches for Tumor Therapy. Cells 2020; 9:E2061. [PMID: 32917034 PMCID: PMC7564019 DOI: 10.3390/cells9092061] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/24/2022] Open
Abstract
Within the last decade, the introduction of checkpoint inhibitors proposed to boost the patients' anti-tumor immune response has proven the efficacy of immunotherapeutic approaches for tumor therapy. Furthermore, especially in the context of the development of biocompatible, cell type targeting nano-carriers, nucleic acid-based drugs aimed to initiate and to enhance anti-tumor responses have come of age. This review intends to provide a comprehensive overview of the current state of the therapeutic use of nucleic acids for cancer treatment on various levels, comprising (i) mRNA and DNA-based vaccines to be expressed by antigen presenting cells evoking sustained anti-tumor T cell responses, (ii) molecular adjuvants, (iii) strategies to inhibit/reprogram tumor-induced regulatory immune cells e.g., by RNA interference (RNAi), (iv) genetically tailored T cells and natural killer cells to directly recognize tumor antigens, and (v) killing of tumor cells, and reprograming of constituents of the tumor microenvironment by gene transfer and RNAi. Aside from further improvements of individual nucleic acid-based drugs, the major perspective for successful cancer therapy will be combination treatments employing conventional regimens as well as immunotherapeutics like checkpoint inhibitors and nucleic acid-based drugs, each acting on several levels to adequately counter-act tumor immune evasion.
Collapse
Affiliation(s)
- Simone Hager
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University (LMU), 81377 Munich, Germany;
| | | | - Ernst Wagner
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University (LMU), 81377 Munich, Germany;
| | - Matthias Bros
- Department of Dermatology, University Medical Center, 55131 Mainz, Germany;
| |
Collapse
|
31
|
Yaseen MM, Abuharfeil NM, Darmani H, Daoud A. Recent advances in myeloid-derived suppressor cell biology. Front Med 2020; 15:232-251. [PMID: 32876877 DOI: 10.1007/s11684-020-0797-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/17/2020] [Indexed: 12/17/2022]
Abstract
In recent years, studying the role of myeloid-derived suppressor cells (MDSCs) in many pathological inflammatory conditions has become a very active research area. Although the role of MDSCs in cancer is relatively well established, their role in non-cancerous pathological conditions remains in its infancy resulting in much confusion. Our objectives in this review are to address some recent advances in MDSC research in order to minimize such confusion and to provide an insight into their function in the context of other diseases. The following topics will be specifically focused upon: (1) definition and characterization of MDSCs; (2) whether all MDSC populations consist of immature cells; (3) technical issues in MDSC isolation, estimation and characterization; (4) the origin of MDSCs and their anatomical distribution in health and disease; (5) mediators of MDSC expansion and accumulation; (6) factors that determine the expansion of one MDSC population over the other; (7) the Yin and Yang roles of MDSCs. Moreover, the functions of MDSCs will be addressed throughout the text.
Collapse
Affiliation(s)
- Mahmoud Mohammad Yaseen
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Nizar Mohammad Abuharfeil
- Department of Biotechnology and Genetic Engineering, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Homa Darmani
- Department of Applied Biology, Faculty of Science and Arts, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Ammar Daoud
- Department of Internal Medicine, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| |
Collapse
|
32
|
Lee YS, Zhang T, Saxena V, Li L, Piao W, Bromberg JS, Scalea JR. Myeloid-derived suppressor cells expand after transplantation and their augmentation increases graft survival. Am J Transplant 2020; 20:2343-2355. [PMID: 32282980 DOI: 10.1111/ajt.15879] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 01/25/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) expand in an inflammatory microenvironment such as cancer and autoimmunity. To study if transplantation induces MDSCs and these cells regulate allograft survival, C57BL/6 donor hearts were transplanted into BALB/c recipients and endogenous MDSCs were characterized. The effects of adoptive transfer of transplant (tx), tumor (tm), and granulocyte-colony stimulating factor (g-csf)-expanded MDSCs or depletion of MDSC were assessed. MDSCs expanded after transplantation (1.7-4.6-fold) in the absence of immunosuppression, homed to allografts, and suppressed proliferation of CD4 T cells in vitro. Tx-MDSCs differed phenotypically from tm-MDSCs and g-csf-MDSCs. Among various surface markers, Rae-1 expression was notably low and TGF-β receptor II was high in tx-MDSCs when compared to tm-MDSCs and g-csf-MDSCs. Adoptive transfer of these three MDSCs led to differential graft survival: control (6 days), tx-MDSCs (7.5 days), tm-MDSCs (9.5 days), and g-csf-MDSCs (19.5 days). In combination with anti-CD154 mAb, MDSCs synergistically extended graft survival from 40 days (anti-CD154 alone) to 86 days with tm-MDSCs and 132 days with g-csf-MDSCs. Early MDSC depletion (day 0 or 20), however, abrogated graft survival, but late depletion (day 25) did not. In conclusion, MDSCs expanded following transplantation, migrated to cardiac allografts, prolonged graft survival, and were synergistic with anti-CD154 mAb.
Collapse
Affiliation(s)
- Young S Lee
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Tianshu Zhang
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vikas Saxena
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Lushen Li
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Wenji Piao
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jonathan S Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Joseph R Scalea
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA.,Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
33
|
Oktem A, Horzum U, Ertop P, Kundakci N, Akay BN, Yalcin B, Esendagli G. Clinical Relevance of Polymorphonuclear Myeloid-Derived Suppressor Cells in Autoimmune-Blistering Disorders Pemphigus Vulgaris and Bullous Pemphigoid. J Invest Dermatol 2020; 141:672-675.e1. [PMID: 32791069 DOI: 10.1016/j.jid.2020.07.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/25/2020] [Accepted: 07/15/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Ayse Oktem
- Department of Dermatology, Faculty of Medicine, Ankara University, Ankara, Turkey.
| | - Utku Horzum
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Pelin Ertop
- Department of Dermatology, Bülent Ecevit University, Zonguldak, Turkey
| | - Nihal Kundakci
- Department of Dermatology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Bengu Nisa Akay
- Department of Dermatology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Basak Yalcin
- Department of Dermatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Gunes Esendagli
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| |
Collapse
|
34
|
Cellular immune dysregulation in the pathogenesis of immune thrombocytopenia. Blood Coagul Fibrinolysis 2020; 31:113-120. [PMID: 31977328 DOI: 10.1097/mbc.0000000000000891] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
: Immune thrombocytopenia (ITP) is an acquired autoimmune hemorrhagic disease characterized by immune-mediated increased platelet destruction and decreased platelet production, resulting from immune intolerance to autoantigen. The pathogenesis of ITP remains unclear, although dysfunction of T and B lymphocytes has been shown to be involved in the pathogenesis of ITP. More recently, it is found that dendritic cells, natural killer, and myeloid-derived suppressor cells also play an important role in ITP. Elucidating its pathogenesis is expected to provide novel channels for the targeted therapy of ITP. This article will review the role of different immune cells in ITP.
Collapse
|
35
|
Iglesias-Escudero M, Sansegundo-Arribas D, Riquelme P, Merino-Fernández D, Guiral-Foz S, Pérez C, Valero R, Ruiz JC, Rodrigo E, Lamadrid-Perojo P, Hutchinson JA, Ochando J, López-Hoyos M. Myeloid-Derived Suppressor Cells in Kidney Transplant Recipients and the Effect of Maintenance Immunotherapy. Front Immunol 2020; 11:643. [PMID: 32425928 PMCID: PMC7203496 DOI: 10.3389/fimmu.2020.00643] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) represent a heterogeneous group of myeloid regulatory cells that were originally described in cancer. Several studies in animal models point to MDSC as important players in the induction of allograft tolerance due to their immune modulatory function. Most of the published studies have been performed in animal models, and the data addressing MDSCs in human organ transplantation are scarce. We evaluated the phenotype and function of different MDSCs subsets in 38 kidney transplant recipients (KTRs) at different time points. Our data indicate that monocytic MDSCs (Mo-MDSC) increase in KTR at 6 and 12 months posttransplantation. On the contrary, the percentages of polymorphonuclear MDSC (PMN-MDSC) and early-stage MDSC (e-MDSC) are not significantly increased. We evaluated the immunosuppressive activity of Mo-MDSC in KTR and confirmed their ability to increase regulatory T cells (Treg) in vitro. Interestingly, when we compared the ability of Mo-MDSC to suppress T cell proliferation, we observed that tacrolimus, but not rapamycin-treated KTR, was able to inhibit CD4+ T cell proliferation in vitro. This indicates that, although mTOR inhibitors are widely regarded as supportive of regulatory responses, rapamycin may impair Mo-MDSC function, and suggests that the choice of immunosuppressive therapy may determine the tolerogenic pathway and participating immune cells that promote organ transplant acceptance in KTR.
Collapse
Affiliation(s)
- María Iglesias-Escudero
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain
| | - David Sansegundo-Arribas
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain.,Department of Immunology, University Hospital Marqués de Valdecilla, Santander, Spain
| | - Paloma Riquelme
- Section of Experimental Surgery, Department of Surgery, University Hospital of Regensburg, Regensburg, Germany
| | - David Merino-Fernández
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain
| | - Sandra Guiral-Foz
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain.,Department of Immunology, University Hospital Marqués de Valdecilla, Santander, Spain
| | - Carmen Pérez
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain
| | - Rosalia Valero
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain.,Department of Nephrology, University Hospital Marqués de Valdecilla, Santander, Spain
| | - Juan Carlos Ruiz
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain.,Department of Nephrology, University Hospital Marqués de Valdecilla, Santander, Spain
| | - Emilio Rodrigo
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain.,Department of Nephrology, University Hospital Marqués de Valdecilla, Santander, Spain
| | - Patricia Lamadrid-Perojo
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain
| | - James A Hutchinson
- Section of Experimental Surgery, Department of Surgery, University Hospital of Regensburg, Regensburg, Germany
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Immunología de Trasplantes, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Marcos López-Hoyos
- Transplantation and Autoimmunity Group, Marqués de Valdecilla Health Research Institute (IDIVAL) Santander, Spain.,Department of Immunology, University Hospital Marqués de Valdecilla, Santander, Spain
| |
Collapse
|
36
|
Trovato R, Canè S, Petrova V, Sartoris S, Ugel S, De Sanctis F. The Engagement Between MDSCs and Metastases: Partners in Crime. Front Oncol 2020; 10:165. [PMID: 32133298 PMCID: PMC7040035 DOI: 10.3389/fonc.2020.00165] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 01/30/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor metastases represent the major cause of cancer-related mortality, confirming the urgent need to identify key molecular pathways and cell-associated networks during the early phases of the metastatic process to develop new strategies to either prevent or control distal cancer spread. Several data revealed the ability of cancer cells to establish a favorable microenvironment, before their arrival in distant organs, by manipulating the cell composition and function of the new host tissue where cancer cells can survive and outgrow. This predetermined environment is termed “pre-metastatic niche” (pMN). pMN development requires that tumor-derived soluble factors, like cytokines, growth-factors and extracellular vesicles, genetically and epigenetically re-program not only resident cells (i.e., fibroblasts) but also non-resident cells such as bone marrow-derived cells. Indeed, by promoting an “emergency” myelopoiesis, cancer cells switch the steady state production of blood cells toward the generation of pro-tumor circulating myeloid cells defined as myeloid-derived suppressor cells (MDSCs) able to sustain tumor growth and dissemination. MDSCs are a heterogeneous subset of myeloid cells with immunosuppressive properties that sustain metastatic process. In this review, we discuss current understandings of how MDSCs shape and promote metastatic dissemination acting in each fundamental steps of cancer progression from primary tumor to metastatic disease.
Collapse
Affiliation(s)
- Rosalinda Trovato
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefania Canè
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Varvara Petrova
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Silvia Sartoris
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Stefano Ugel
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| | - Francesco De Sanctis
- Section of Immunology, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
37
|
Islam J, Lee HJ, Yang SH, Kim DK, Joo KW, Kim YS, Seo SU, Seong SY, Lee DS, Youn JI, Han SS. Expansion of Myeloid-Derived Suppressor Cells Correlates with Renal Progression in Type 2 Diabetic Nephropathy. Immune Netw 2020; 20:e18. [PMID: 32395370 PMCID: PMC7192828 DOI: 10.4110/in.2020.20.e18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/27/2020] [Accepted: 02/06/2020] [Indexed: 01/04/2023] Open
Abstract
Type 2 diabetic nephropathy (T2DN) progresses with an increasingly inflammatory milieu, wherein various immune cells are relevant. Herein, we investigated the levels of myeloid-derived suppressor cells (MDSCs) and their clinical implication in patients with T2DN. A total of 91 subjects (T2DN, n=80; healthy, n=11) were recruited and their PBMCs were used for flow cytometric analysis of polymorphonuclear (PMN-) and monocytic (M-) MDSCs, in addition to other immune cell subsets. The risk of renal progression was evaluated according to the quartiles of MDSC levels using the Cox model. The proportion of MDSCs in T2DN patients was higher than in healthy individuals (median, 6.7% vs. 2.5%). PMN-MDSCs accounted for 96% of MDSCs, and 78% of PMN-MDSCs expressed Lox-1. The expansion of PMN-MDSCs was not related to the stage of T2DN or other kidney disease parameters such as glomerular filtration rate and proteinuria. The production of ROS in PMN-MDSCs of patients was higher than in neutrophils of patients or in immune cells of healthy individuals, and this production was augmented under hyperglycemic conditions. The 4th quartile group of PMN-MDSCs had a higher risk of renal progression than the 1st quartile group, irrespective of adjusting for multiple clinical and laboratory variables. In conclusion, PMN-MDSCs are expanded in patients with T2DN, and may represent as an immunological biomarker of renal progression.
Collapse
Affiliation(s)
- Jahirul Islam
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hack June Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seung Hee Yang
- Kidney Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Kwon Wook Joo
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Yon Su Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Sang-Uk Seo
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon 25159, Korea
| | - Seung-Yong Seong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon 25159, Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Dong-Sup Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Je-In Youn
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Wide River Institute of Immunology, Seoul National University College of Medicine, Hongcheon 25159, Korea.,Research Institute, ProGen Inc., Seongnam 13488, Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea.,Kidney Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
38
|
Khorooshi R, Marczynska J, Dieu RS, Wais V, Hansen CR, Kavan S, Thomassen M, Burton M, Kruse T, Webster GA, Owens T. Innate signaling within the central nervous system recruits protective neutrophils. Acta Neuropathol Commun 2020; 8:2. [PMID: 31915070 PMCID: PMC6950927 DOI: 10.1186/s40478-019-0876-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/20/2019] [Indexed: 01/27/2023] Open
Abstract
There is great interest in understanding how the central nervous system (CNS) communicates with the immune system for recruitment of protective responses. Infiltrating phagocytic monocytes and granulocytes are implicated in neuroinflammation in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). To investigate how CNS endogenous signals can be harnessed to promote anti-inflammatory programs, we have used a particulate Toll-like receptor 9 and nucleotide-oligomerization domain 2 bispecific innate ligand (MIS416), to address whether its phagocytosis within the CNS recruits protective myeloid cells. We find that MIS416 injected intrathecally into the cerebrospinal fluid via the cisterna magna induced a local chemokine response that recruited blood-derived monocytes and neutrophils to the CNS. These cells phagocytosed MIS416. The increase in EAE severity normally seen from time of onset did not occur in mice receiving MIS416. This suppression of disease symptoms was dependent on expression of the type I interferon receptor (IFNAR). Transfer of intrathecal MIS416-induced neutrophils suppressed EAE in recipient mice, while monocytes did not transfer protection. MIS416-induced neutrophils showed increased IL-10 expression that was IFNAR1-driven. In contrast to intrathecal administration, intravenous administration of MIS416 led to monocyte but not neutrophil infiltration to the CNS. We thus identify a CNS-intrinsic and -specific phagocytosis-induced recruitment of anti-inflammatory neutrophils that contribute to CNS homeostasis and may have therapeutic potential.
Collapse
|
39
|
Dysthe M, Parihar R. Myeloid-Derived Suppressor Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1224:117-140. [DOI: 10.1007/978-3-030-35723-8_8] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
40
|
Wang B, Dong G, Zhang Q, Yan F, Li Z, Li C, Zhang H, Ma Q, Dai J, Si C, Xiong H. The inhibitor of autophagy SBI-0206965 aggravates atherosclerosis through decreasing myeloid-derived suppressor cells. Exp Ther Med 2019; 19:1370-1378. [PMID: 32010311 PMCID: PMC6966176 DOI: 10.3892/etm.2019.8317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis (AS) is currently the leading cause of mortality worldwide, with the development of new strategies to prevent the formation and rupture of atherosclerotic plaques being a paramount area of research. Amounting evidence suggests autophagy has an important role in the pathogenesis of AS and may be a potential therapeutic target. In this study, the effect of SBI-0206965(6965), a novel inhibitor of autophagy, was tested on the development of AS in apolipoprotein E deficient (ApoE−/−) mice. Systemic application of 6965 was found to aggravate AS, with increased plaque size and decreased plaque stability in comparison with the control. Of note, it was observed that 6965 decreased the proportion of myeloid-derived suppressor cells (MDSCs). Further investigation demonstrated MDSCs markedly alleviated AS in ApoE−/− mice; while 6965 reduced the viability and promoted apoptosis of MDSCs in vitro. This is the first study describing an association between autophagy and MDSCs in AS models, providing a novel mechanism to potentially target in the management of this condition.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China.,Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Guanjun Dong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Qingqiing Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Fenglian Yan
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Zhihua Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Chunxia Li
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Qun Ma
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Jun Dai
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Chuanping Si
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, Shandong 272067, P.R. China
| | - Huabao Xiong
- Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
41
|
Agrati C, Tumino N, Bordoni V, Pinnetti C, Sabatini A, Amendola A, Abbate I, Lorenzini P, Mondi A, Casetti R, Cimini E, Grassi G, Antinori A, Sacchi A. Myeloid Derived Suppressor Cells Expansion Persists After Early ART and May Affect CD4 T Cell Recovery. Front Immunol 2019; 10:1886. [PMID: 31440256 PMCID: PMC6694843 DOI: 10.3389/fimmu.2019.01886] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/25/2019] [Indexed: 12/26/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSC) are expanded during HIV-1 infection and correlated with disease progression. MDSC expand in the early phase of primary infection depending on TRAIL level. In this study we evaluated the effect of ART on the frequency of MDSC in patients with primary HIV infection (PHI), and their impact on CD4 T cell reconstitution. MDSC frequency was evaluated by flow-cytometry in 60 PHI patients at 12, 24 and 48 weeks after ART initiation. Cytokine plasma levels were evaluated by Luminex technology at the same time points. The capacity of MDSC to modulate hematopoietic early progenitor cells' expansion was evaluated using the OP9/Dl1 in vitro system. As previously described, polymorphonuclear-MDSC (PMN-MDSC) frequency was higher in PHI compared to healthy donors. Interestingly, 48 weeks of successful ART failed to normalize the PMN-MDSC frequency. Moreover, PMN-MDSC frequency was not correlated with residual viral load, suggesting that the persistence of PMN-MDSC was not due to residual viral replication. Interestingly, patients with low PMN-MDSC frequency (<6%) at T0 had a higher HIV DNA at the same time point than individuals with high PMN-MDSC frequency (>6%). We also found an inverse correlation between PMN-MDSC frequency and CD4-T cell count at 48 weeks post-ART, which was confirmed by multivariate analysis adjusting for age and CD4 T cell number at baseline. These data suggest that the persistence of PMN-MDSC may impact CD4 T cell recovery. Indeed, in vitro PMN-MDSC impaired the expansion of CD34+CD38- hematopoietic early progenitors. Further, a balance between TRAIL and GM-CSF may be necessary to maintain a low MDSC level. In conclusion, early ART initiation was not able to normalize PMN-MDSC frequency that might impact the CD4 T cell recovery. These data open new questions regarding the clinical impact of MDSC persistence in HIV+ patients, in particular on non-AIDS related diseases.
Collapse
Affiliation(s)
- Chiara Agrati
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Nicola Tumino
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Veronica Bordoni
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Carmela Pinnetti
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Sabatini
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Alessandra Amendola
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Isabella Abbate
- Virology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Patrizia Lorenzini
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Annalisa Mondi
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Rita Casetti
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Germana Grassi
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Andrea Antinori
- Clinical Division, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| | - Alessandra Sacchi
- Cellular Immunology Laboratory, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, Rome, Italy
| |
Collapse
|
42
|
Pawelec G, Verschoor CP, Ostrand-Rosenberg S. Myeloid-Derived Suppressor Cells: Not Only in Tumor Immunity. Front Immunol 2019; 10:1099. [PMID: 31156644 PMCID: PMC6529572 DOI: 10.3389/fimmu.2019.01099] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Since the realization that immature myeloid cells are powerful modulators of the immune response, many studies on “myeloid-derived suppressor cells” (MDSCs) have documented their ability to promote tumor progression in melanoma and other cancers. Whether MDSCs are induced solely pathologically in tumorigenesis, or whether they also represent physiological immune control mechanisms, is not well-understood, but is particularly important in the light of ongoing attempts to block their activities in order to enhance anti-tumor immunity. Here, we briefly review studies which explore (1) how best to identify MDSCs in the context of cancer and how this compares to other conditions in humans; (2) what the suppressive mechanisms of MDSCs are and how to target them pharmacologically; (3) whether levels of MDSCs with various phenotypes are informative for clinical outcome not only in cancer but also other diseases, and (4) whether MDSCs are only found under pathological conditions or whether they also represent a physiological regulatory mechanism for the feedback control of immunity. Studies unequivocally document that MDSCs strongly influence cancer outcomes, but are less informative regarding their relevance to infection, autoimmunity, transplantation and aging, especially in humans. So far, the results of clinical interventions to reverse their negative effects in cancer have been disappointing; thus, developing differential approaches to modulate MSDCs in cancer and other diseases without unduly comprising any normal physiological function requires further exploration.
Collapse
Affiliation(s)
- Graham Pawelec
- Department of Immunology, University of Tübingen, Tübingen, Germany.,Health Sciences North Research Institute, Sudbury, ON, Canada
| | - Chris P Verschoor
- Health Sciences North Research Institute, Sudbury, ON, Canada.,Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Suzanne Ostrand-Rosenberg
- Department of Pathology and Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
43
|
Cho ME, Branton MH, Smith DA, Bartlett L, Howard L, Reynolds JC, Rosenstein D, Sethi S, Nava MB, Barisoni L, Fervenza FC, Kopp JB. Open-Label Clinical Trials of Oral Pulse Dexamethasone for Adults with Idiopathic Nephrotic Syndrome. Am J Nephrol 2019; 49:377-385. [PMID: 30965344 DOI: 10.1159/000497064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/11/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND In adults with primary focal segmental glomerulosclerosis (FSGS), daily prednisone may induce complete remissions (CR) and partial remissions (PR), but relapses are frequent and adverse events are common. METHODS We carried out 2 open-label, uncontrolled trials to explore the efficacy and tolerability of pulse oral dexamethasone as an alternative to daily prednisone. We enrolled adult patients with proteinuria > 3.5 g/day despite the use of renin-angiotensin-aldosterone blockade. In the first trial, we enrolled 14 subjects with FSGS and administered 4 dexamethasone doses (25 mg/m2) daily for 4 days, repeated every 28 days over 32 weeks. The second trial involved a more intensive regimen. Eight subjects received 4 dexamethasone doses of 50 mg/m2 every 4 weeks for 12 weeks, followed by 4 doses of 25 mg/m2 every 4 weeks for 36 weeks; subjects were randomized to 2 doses every 2 weeks or 4 doses every 4 weeks. RESULTS In the first trial, we enrolled 13 subjects with FSGS and 1 with minimal change disease and found a combined CR and PR rate of 36%. In the second trial, we enrolled 8 subjects. The combined CR and PR rate was 29%. Analysis combining both trials showed a combined CR and PR rate of 33%. Adverse events were observed in 32% of subjects, with mood symptoms being most common. There were no serious adverse events related to the study. CONCLUSION We conclude that high dose oral dexamethasone is well tolerated by adults with idiopathic nephrotic syndrome and may have some efficacy.
Collapse
Affiliation(s)
- Monique E Cho
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, Maryland, USA
| | - Mary H Branton
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, Maryland, USA
| | - David A Smith
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, Maryland, USA
| | - Linda Bartlett
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, Maryland, USA
| | - Lilian Howard
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, Maryland, USA
| | - James C Reynolds
- Radiology and Imaging Services, Mark O. Hatfield Clinical Research Center, Bethesda, Maryland, USA
| | - Donald Rosenstein
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Sanjeev Sethi
- Department of Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - M Berenice Nava
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, Maryland, USA
- Kaiser Permanente, San Diego, California, USA
| | - Laura Barisoni
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Fernando C Fervenza
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Disease, Bethesda, Maryland, USA,
| |
Collapse
|
44
|
Mu W, Wang Z, Zöller M. Ping-Pong-Tumor and Host in Pancreatic Cancer Progression. Front Oncol 2019; 9:1359. [PMID: 31921628 PMCID: PMC6927459 DOI: 10.3389/fonc.2019.01359] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Metastasis is the main cause of high pancreatic cancer (PaCa) mortality and trials dampening PaCa mortality rates are not satisfying. Tumor progression is driven by the crosstalk between tumor cells, predominantly cancer-initiating cells (CIC), and surrounding cells and tissues as well as distant organs, where tumor-derived extracellular vesicles (TEX) are of major importance. A strong stroma reaction, recruitment of immunosuppressive leukocytes, perineural invasion, and early spread toward the peritoneal cavity, liver, and lung are shared with several epithelial cell-derived cancer, but are most prominent in PaCa. Here, we report on the state of knowledge on the PaCIC markers Tspan8, alpha6beta4, CD44v6, CXCR4, LRP5/6, LRG5, claudin7, EpCAM, and CD133, which all, but at different steps, are engaged in the metastatic cascade, frequently via PaCIC-TEX. This includes the contribution of PaCIC markers to TEX biogenesis, targeting, and uptake. We then discuss PaCa-selective features, where feedback loops between stromal elements and tumor cells, including distorted transcription, signal transduction, and metabolic shifts, establish vicious circles. For the latter particularly pancreatic stellate cells (PSC) are responsible, furnishing PaCa to cope with poor angiogenesis-promoted hypoxia by metabolic shifts and direct nutrient transfer via vesicles. Furthermore, nerves including Schwann cells deliver a large range of tumor cell attracting factors and Schwann cells additionally support PaCa cell survival by signaling receptor binding. PSC, tumor-associated macrophages, and components of the dysplastic stroma contribute to perineural invasion with signaling pathway activation including the cholinergic system. Last, PaCa aggressiveness is strongly assisted by the immune system. Although rich in immune cells, only immunosuppressive cells and factors are recovered in proximity to tumor cells and hamper effector immune cells entering the tumor stroma. Besides a paucity of immunostimulatory factors and receptors, immunosuppressive cytokines, myeloid-derived suppressor cells, regulatory T-cells, and M2 macrophages as well as PSC actively inhibit effector cell activation. This accounts for NK cells of the non-adaptive and cytotoxic T-cells of the adaptive immune system. We anticipate further deciphering the molecular background of these recently unraveled intermingled phenomena may turn most lethal PaCa into a curatively treatable disease.
Collapse
Affiliation(s)
- Wei Mu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Wei Mu
| | - Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong, Pharmaceutical University, Guangzhou, China
| | - Margot Zöller
- Department of Oncology, The First Affiliated Hospital of Guangdong, Pharmaceutical University, Guangzhou, China
| |
Collapse
|
45
|
Budhwar S, Verma P, Verma R, Rai S, Singh K. The Yin and Yang of Myeloid Derived Suppressor Cells. Front Immunol 2018; 9:2776. [PMID: 30555467 PMCID: PMC6280921 DOI: 10.3389/fimmu.2018.02776] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
In recent years, most of our knowledge about myeloid derived suppressor cells (MDSCs) has come from cancer studies, which depicts Yin side of MDSCs. In cancer, inherent immunosuppressive action of MDSCs favors tumor progression by inhibiting antitumor immune response. However, recently Yang side of MDSCs has also been worked out and suggests the role in maintenance of homeostasis during non-cancer situations like pregnancy, obesity, diabetes, and autoimmune disorders. Continued work in this area has armored the biological importance of these cells as master regulators of immune system and prompted scientists all over the world to look from a different perspective. Therefore, explicating Yin and Yang arms of MDSCs is obligatory to use it as a double edged sword in a much smarter way. This review is an attempt toward presenting a synergistic coalition of all the facts and controversies that exist in understanding MDSCs, bring them on the same platform and approach their "Yin and Yang" nature in a more comprehensive and coherent manner.
Collapse
Affiliation(s)
- Snehil Budhwar
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Priyanka Verma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Rachna Verma
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Sangeeta Rai
- Department of Obstetrics and Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Kiran Singh
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
46
|
Sacchi A, Tumino N, Grassi G, Casetti R, Cimini E, Bordoni V, Ammassari A, Antinori A, Agrati C. A new procedure to analyze polymorphonuclear myeloid derived suppressor cells in cryopreserved samples cells by flow cytometry. PLoS One 2018; 13:e0202920. [PMID: 30161175 PMCID: PMC6117014 DOI: 10.1371/journal.pone.0202920] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 08/10/2018] [Indexed: 12/20/2022] Open
Abstract
Myeloid derived suppressor cells (MDSC) is a heterogeneous subset of immature and mature cells of the myeloid lineage, undergoing expansion during pathologic conditions, and able to perform strong immune suppressive functions. It has been shown that cryopreservation selectively impacts the polimorphonuclear (PMN) MDSC viability and recovery, and alters the correct analysis of MDSC subsets. In laboratory practice, cryopreservation is often inevitable, in particular in multicenter studies where samples have to be shipped to a centralized laboratory. Aim of the present work was to set out a new protocol to evaluate the frequency of PMN-MDSC in thawed cells by flow-cytometry. PBMC were isolated from HIV+ patients and healthy donors, and were cryopreserved for at least ten days. After thawing, two different protocols were used: 1. standard protocol (SP) consisting of staining with the antibodies mix and then fixing with formalin 1%; 2. thawed protocol (TP) in which fixation foregoes the staining with the antibodies mix. Results showed that PMN-MDSC frequency in ex vivo PBMC evaluated by means TP was comparable to that analysed by SP, indicating that the protocol did not alter PMN-MDSC quantification in ex vivo cells. We then demonstrated that PMN-MDSC frequency in thawed PBMC tested by TP was almost identical to the frequency obtained in ex vivo cells evaluated by using SP. However, we observed that after three hours of culture post-thawing, PMN-MDSC were not assessable anymore with both SP and TP. In conclusion, we herein demonstrated that fixing PBMC soon after thawing and before antibody staining allows preservation of PMN-MDSC integrity and a reliable cells quantification. Thus, it is possible to phenotipically identify PMN-MDSC in cryopreserved PBMC, consenting adequate test precision and accuracy as well as making multicentre research more feasible.
Collapse
Affiliation(s)
- Alessandra Sacchi
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
- * E-mail:
| | - Nicola Tumino
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Germana Grassi
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Rita Casetti
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Eleonora Cimini
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Veronica Bordoni
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Adriana Ammassari
- Clinical Department, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Andrea Antinori
- Clinical Department, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| | - Chiara Agrati
- Cellular Immunology and Pharmacology Laboratory, “Lazzaro Spallanzani” National Institute for Infectious Diseases, IRCCS, Rome, Italy
| |
Collapse
|
47
|
Schep S, Schutgens R, Fischer K, Boes M. Review of immune tolerance induction in hemophilia A. Blood Rev 2018; 32:326-338. [DOI: 10.1016/j.blre.2018.02.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/01/2018] [Accepted: 02/13/2018] [Indexed: 12/22/2022]
|
48
|
The Expansion of Myeloid-Derived Suppressor Cells Is Associated with Joint Inflammation in Rheumatic Patients with Arthritis. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5474828. [PMID: 30046600 PMCID: PMC6038685 DOI: 10.1155/2018/5474828] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/17/2018] [Accepted: 05/10/2018] [Indexed: 12/23/2022]
Abstract
Introduction We investigated the proportion of myeloid-derived suppressor cells (MDSCs) and their subsets in patients with rheumatic diseases and clarified the association between these cells and the patient clinical data. Methods Patients with rheumatic diseases and healthy controls were recruited. The clinical characteristics were obtained. The MDSCs and their subsets were marked with fluorescently labelled antibodies and were then analyzed with flow cytometry. Results The patients included 31 with RA, 21 with AS, 14 with OA, 11 with SLE with arthritis, 13 with SLE without arthritis, 9 with Gout, 10 with HUA, and 25 healthy controls. The proportions of MDSCs, M-MDSCs, and G-MDSCs were higher in patients with RA than in healthy controls (6.56±6.77% versus 1.46±0.96%, 2.52±3.81% versus 0.35±0.35%, and 1.13±1.64% versus 0.18±0.14%; p<0.001). The same increased cells were also found in other patients. The proportions of MDSCs and M-MDSCs were mostly correlated with the patient's joint inflammation indexes and the disease activity. When other cell subsets were adjusted, the increased risk of arthritis was also obtained for M-MDSCs (adjusted OR=5.772; p=0.031). Conclusions The expansion of MDSCs and their subsets was correlated with the disease activity and joint inflammation in patient with different rheumatic diseases. The proportion of M-MDSCs was associated with the risk of arthritis in those populations.
Collapse
|
49
|
Kondoh N, Mizuno-Kamiya M, Takayama E, Kawati H, Umemura N, Yamazaki Y, Mitsudo K, Tohnai I. Perspectives of Immune Suppression in the Tumor Microenvironment Promoting Oral Malignancy. Open Dent J 2018; 12:455-465. [PMID: 29988281 PMCID: PMC6018133 DOI: 10.2174/1874210601812010455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/15/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023] Open
Abstract
Introduction In order to survive, cancers control immune systems and evade immune detection using mediators consisting of immune checkpoint molecules and cellular systems associated with immune suppression. Methodology During the development of cancer and chronic infections, the immune checkpoints and cellular components including regulatory T cells, myeloid derived suppressor cells and cancer associated fibroblasts are often enhanced as a mechanism of immune subversion and have therefore become very important therapeutic targets. Conclusion In this review, we will discuss the complexity of immune-suppressive mechanisms in the tumor milieu of cancers, including oral malignancy.
Collapse
Affiliation(s)
- Nobuo Kondoh
- Department of Oral Biochemistry, Division of Oral Structure, Function and Development, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Masako Mizuno-Kamiya
- Department of Management and Information Studies, Chemistry Laboratory, Asahi University School of Business Administration, 1851 Hozumi, Mizuho, Gifu 501-0296, Japan
| | - Eiji Takayama
- Department of Oral Biochemistry, Division of Oral Structure, Function and Development, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Harumi Kawati
- Department of Oral Biochemistry, Division of Oral Structure, Function and Development, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Naoki Umemura
- Department of Oral Biochemistry, Division of Oral Structure, Function and Development, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Yutaka Yamazaki
- Department of Oral Health Science, Gerodontology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Kita-13, Nishi-7, Kita-ku, Sapporo, 060-8586, Japan
| | - Kenji Mitsudo
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Iwai Tohnai
- Department of Oral and Maxillofacial Surgery, Yokohama City University Graduate School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| |
Collapse
|
50
|
Zöller M, Zhao K, Kutlu N, Bauer N, Provaznik J, Hackert T, Schnölzer M. Immunoregulatory Effects of Myeloid-Derived Suppressor Cell Exosomes in Mouse Model of Autoimmune Alopecia Areata. Front Immunol 2018; 9:1279. [PMID: 29951053 PMCID: PMC6008552 DOI: 10.3389/fimmu.2018.01279] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 05/22/2018] [Indexed: 01/28/2023] Open
Abstract
The treatment of autoimmune diseases still poses a major challenge, frequently relying on non-specific immunosuppressive drugs. Current efforts aim at reestablishing self tolerance using immune cells with suppressive activity like the regulatory T cells (Treg) or the myeloid-derived suppressor cells (MDSC). We have demonstrated therapeutic efficacy of MDSC in mouse Alopecia Areata (AA). In the same AA model, we now asked whether MDSC exosomes (MDSC-Exo) can replace MDSC. MDSC-Exo from bone marrow cells (BMC) cultures of healthy donors could substantially facilitate treatment. With knowledge on MDSC-Exo being limited, their suitability needs to be verified in advance. Protein marker profiles suggest comparability of BMC- to ex vivo collected inflammatory MDSC/MDSC-Exo in mice with a chronic contact dermatitis, which is a therapeutic option in AA. Proteome analyses substantiated a large overlap of function-relevant molecules in MDSC and MDSC-Exo. Furthermore, MDSC-Exo are taken up by T cells, macrophages, NK, and most avidly by Treg and MDSC-Exo uptake exceeds binding of MDSC themselves. In AA mice, MDSC-Exo preferentially target skin-draining lymph nodes and cells in the vicinity of remnant hair follicles. MDSC-Exo uptake is accompanied by a strong increase in Treg, reduced T helper proliferation, mitigated cytotoxic activity, and a slight increase in lymphocyte apoptosis. Repeated MDSC-Exo application in florid AA prevented progression and sufficed for partial hair regrowth. Deep sequencing of lymphocyte mRNA from these mice revealed a significant increase in immunoregulatory mRNA, including FoxP3 and arginase 1. Downregulated mRNA was preferentially engaged in prohibiting T cell hyperreactivity. Taken together, proteome analysis provided important insights into potential MDSC-Exo activities, these Exo preferentially homing into AA-affected organs. Most importantly, changes in leukocyte mRNA seen after treatment of AA mice with MDSC-Exo sustainably supports the strong impact on the adaptive and the non-adaptive immune system, with Treg expansion being a dominant feature. Thus, MDSC-Exo could potentially serve as therapeutic agents in treating AA and other autoimmune diseases.
Collapse
Affiliation(s)
- Margot Zöller
- Tumor Cell Biology, Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Kun Zhao
- Tumor Cell Biology, Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Natalia Kutlu
- Tumor Cell Biology, Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Nathalie Bauer
- Tumor Cell Biology, Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Jan Provaznik
- Gene Core Unit, EMBL Heidelberg, Heidelberg, Germany
| | - Thilo Hackert
- Pancreas Section, Department of Surgery, University Hospital of Heidelberg, Heidelberg, Germany
| | - Martina Schnölzer
- Functional Proteome Analysis, German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|