1
|
Aalinkeel R, Quigg RJ, Alexander J. The complement system and kidney cancer: pathogenesis to clinical applications. J Clin Invest 2025; 135:e188351. [PMID: 40309765 PMCID: PMC12043091 DOI: 10.1172/jci188351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Kidney cancer poses unique clinical challenges because of its resistance to conventional treatments and its tendency to metastasize. The kidney is particularly susceptible to dysfunction of the complement system, an immune network that tumors often exploit. Recent discoveries have highlighted that the complement system not only plays a crucial role in immune surveillance and defense in the circulatory system, but also functions intracellularly and autonomously. This concept has shifted the focus of investigation toward understanding how complement proteins influence cancer progression by regulating the tumor microenvironment (TME), cell signaling, proliferation, metabolism, and the immune response. With the complement system and its inhibitors emerging as a promising new class of immunotherapeutics and potential complement-targeted treatments advancing through development pipelines and clinical trials, this Review provides a timely examination of how harnessing the complement system could lead to effective tumor treatments and how to strategically combine complement inhibitors with other cancer treatments, offering renewed hope in the fight against kidney cancer.
Collapse
|
2
|
Xu X, Zhang S, Luo Z, Zheng Y, Kong T, Huang C, Qiu Z. Frontiers and Controversies in De Novo Gastrointestinal Tumors After Organ Transplantation: Current Progress and Future Directions. Ann Surg Oncol 2025; 32:3392-3405. [PMID: 40035907 DOI: 10.1245/s10434-025-16975-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025]
Abstract
The increasing success of organ transplantation has significantly improved survival for patients with end-stage diseases, yet it introduces a complex dilemma: the elevated risk for the development of de novo gastrointestinal (GI) tumors. The sustained immunosuppression required to maintain graft function paradoxically undermines the body's natural defenses against cancer, leading to a higher incidence, aggressive progression, and atypical presentations of GI tumors among transplant recipients compared with the general population. This presents a pressing challenge: balancing the dual imperatives of preventing graft rejection and effectively managing malignancies. Current treatment paradigms, including surgical approaches, chemotherapy, radiation therapy, and the emerging role of immunotherapy, are fraught with complexities due to the altered immune landscape in these patients. This review underscores the critical need to understand the multifaceted relationship between post-transplantation immunosuppression and tumorigenesis, providing a comprehensive exploration of epidemiologic shifts, pathophysiologic insights, and the intricacies of the tumor microenvironment in this unique patient population. Understanding and managing GI tumors in transplant recipients is not only a clinical challenge, but also a necessary frontier in transplant oncology, promising to refine therapeutic strategies and improve the longevity and quality of life for this growing patient cohort.
Collapse
Affiliation(s)
- Ximo Xu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaopeng Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zai Luo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Zheng
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Kong
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhengjun Qiu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Cui X, Song Y, Han J, Yuan Z. The multifaceted role of SMAD4 in immune cell function. Biochem Biophys Rep 2025; 41:101902. [PMID: 39802394 PMCID: PMC11721226 DOI: 10.1016/j.bbrep.2024.101902] [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: 08/30/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
The Transforming Growth Factor-beta (TGF-β) signaling pathway, with SMAD4 as its central mediator, plays a pivotal role in regulating cellular functions, including growth, differentiation, apoptosis, and immune responses. While extensive research has elucidated SMAD4's role in tumorigenesis, its functions within immune cells remain underexplored. This review synthesizes current knowledge on SMAD4's diverse roles in various immune cells such as T cells, B cells, dendritic cells, and macrophages, highlighting its impact on immune homeostasis and pathogen response. Understanding SMAD4's role in immune cells is crucial, as its dysregulation can lead to autoimmune disorders, chronic inflammation, and immune deficiencies. The review emphasizes the significance of SMAD4 in immune regulation, proposing that deeper investigation could reveal novel therapeutic targets for immune-mediated conditions. Insights into SMAD4's involvement in processes like T cell differentiation, B cell class switch recombination, and macrophage polarization underscore its potential as a therapeutic target for a range of diseases, including autoimmune disorders and cancer.
Collapse
Affiliation(s)
- Xinmu Cui
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Yu Song
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| | - Jianfeng Han
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
- Cellular Biomedicine Group Inc, Shanghai, 201203, China
| | - Zhaoxin Yuan
- Changchun Medical College, 6177, Jilin Street, Changchun, 130031, China
| |
Collapse
|
4
|
DeBerge M, Schroth S, Du F, Yeap XY, Wang JJ, Zhang ZJ, Ansari MJ, Scott EA, Thorp EB. Hypoxia inducible factor 2α promotes tolerogenic macrophage development during cardiac transplantation through transcriptional regulation of colony stimulating factor 1 receptor. Proc Natl Acad Sci U S A 2024; 121:e2319623121. [PMID: 38889142 PMCID: PMC11214057 DOI: 10.1073/pnas.2319623121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
Solid organ transplantation mobilizes myeloid cells, including monocytes and macrophages, which are central protagonists of allograft rejection. However, myeloid cells can also be functionally reprogrammed by perioperative costimulatory blockade to promote a state of transplantation tolerance. Transplantation tolerance holds promise to reduce complications from chronic immunosuppression and promote long-term survival in transplant recipients. We sought to identify different mediators of transplantation tolerance by performing single-cell RNA sequencing of acute rejecting or tolerized cardiac allografts. This led to the unbiased identification of the transcription factor, hypoxia inducible factor (HIF)-2α, in a subset of tolerogenic monocytes. Using flow cytometric analyses and mice with conditional loss or gain of function, we uncovered that myeloid cell expression of HIF-2α was required for costimulatory blockade-induced transplantation tolerance. While HIF-2α was dispensable for mobilization of tolerogenic monocytes, which were sourced in part from the spleen, it promoted the expression of colony stimulating factor 1 receptor (CSF1R). CSF1R mediates monocyte differentiation into tolerogenic macrophages and was found to be a direct transcriptional target of HIF-2α in splenic monocytes. Administration of the HIF stabilizer, roxadustat, within micelles to target myeloid cells, increased HIF-2α in splenic monocytes, which was associated with increased CSF1R expression and enhanced cardiac allograft survival. These data support further exploration of HIF-2α activation in myeloid cells as a therapeutic strategy for transplantation tolerance.
Collapse
Affiliation(s)
- Matthew DeBerge
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
- Department of Anesthesiology, Critical Care and Pain Medicine, University of Texas Health Science Center, Houston, TX77030
| | - Samantha Schroth
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Fanfan Du
- Department of Biomedical Engineering, Northwestern University, Evanston, IL60208
| | - Xin Yi Yeap
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Jiao-Jing Wang
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, IL60611
| | - Zheng Jenny Zhang
- Department of Surgery, Comprehensive Transplant Center, Northwestern University, Chicago, IL60611
| | - Mohammed Javeed Ansari
- Division of Nephrology and Hypertension, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL60208
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| |
Collapse
|
5
|
Mathew JM, Sanders JM, Cirocco R, Miller J, Leventhal JR. Differentiation of regulatory myeloid and T-cells from adult human hematopoietic stem cells after allogeneic stimulation. Front Immunol 2024; 15:1366972. [PMID: 38455047 PMCID: PMC10918006 DOI: 10.3389/fimmu.2024.1366972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction Donor hematopoietic stem cell (DHSC) infusions are increasingly being studied in transplant patients for tolerance induction. Methods To analyze the fate of infused DHSCs in patients, we developed an in vitro culture system utilizing CD34+DHSCs stimulated with irradiated allogeneic cells in cytokine supplemented medium long-term. Results Flow cytometric analyses revealed loss of the CD34 marker and an increase in CD33+ myeloid and CD3+ T-cell proportion by 10.4% and 72.7%, respectively, after 21 days in culture. T-cells primarily expressed TcR-αβ and were of both CD4+ and CD8+ subsets. Approximately 80% of CD3+ T cells lacked expression of the co-stimulatory receptor CD28. The CD4+ compartment was predominated by CD4+CD25+CD127-FOXP3+ Tregs (>50% CD4+CD127- compartment) with <1% of all leukocytes exhibiting a CD4+CD127+ phenotype. Molecular analyses for T-cell receptor excision circles showed recent and increased numbers of TcR rearrangements in generated T cells over time suggesting de novo differentiation from DHSCs. CD33+ myeloid cells mostly expressed HLA-DR, but lacked expression of co-stimulatory receptors CD80 and CD83. When studied as modulators in primary mixed lymphocyte reactions where the cells used to stimulate the DHSC were used as responders, the DHSC-lines and their purified CD8+, CD4+, CD33+ and linage negative subsets inhibited the responses in a dose-dependent and non-specific fashion. The CD8+ cell-mediated inhibition was due to direct lysis of responder cells. Discussion Extrapolation of these results into the clinical situation would suggest that DHSC infusions into transplant recipients may generate multiple subsets of donor "chimeric" cells and promote recipient Treg development that could regulate the anti-donor immune response in the periphery. These studies have also indicated that T cell maturation can occur in vitro in response to allogeneic stimulation without the pre-requisite of a thymic-like environment or NOTCH signaling stimulatory cell line.
Collapse
Affiliation(s)
- James M. Mathew
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Jes M. Sanders
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Robert Cirocco
- HLA Laboratory, LeHigh Valley Health Network, Allentown, PA, United States
| | - Joshua Miller
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| | - Joseph R. Leventhal
- Comprehensive Transplant Center, Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, United States
| |
Collapse
|
6
|
Bhat AA, Afzal O, Afzal M, Gupta G, Thapa R, Ali H, Hassan Almalki W, Kazmi I, Alzarea SI, Saleem S, Samuel VP, Gubbiyappa SK, Subramaniyan V. MALAT1: A key regulator in lung cancer pathogenesis and therapeutic targeting. Pathol Res Pract 2024; 253:154991. [PMID: 38070223 DOI: 10.1016/j.prp.2023.154991] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 11/21/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024]
Abstract
Lung cancer remains a formidable global health burden, necessitating a comprehensive understanding of the underlying molecular mechanisms driving its progression. Recently, lncRNAs have become necessary controllers of various biological functions, including cancer development. MALAT1 has garnered significant attention due to its multifaceted role in lung cancer progression. Lung cancer, among other malignancies, upregulates MALAT1. Its overexpression has been associated with aggressive tumor behavior and poor patient prognosis. MALAT1 promotes cellular proliferation, epithelial-mesenchymal transition (EMT), and angiogenesis in lung cancer, collectively facilitating tumor growth and metastasis. Additionally, MALAT1 enhances cancer cell invasion by interacting with numerous signaling pathways. Furthermore, MALAT1 has been implicated in mediating drug resistance in lung cancer, contributing to the limited efficacy of conventional therapies. Recent advancements in molecular biology and high-throughput sequencing technologies have offered fresh perspectives into the regulatory networks of MALAT1 in lung cancer. It exerts its oncogenic effects by acting as a ceRNA to sponge microRNAs, thereby relieving their inhibitory effects on target genes. Moreover, MALAT1 also influences chromatin remodeling and post-translational modifications to modulate gene expression, further expanding its regulatory capabilities. This review sheds light on the multifaceted roles of MALAT1 in lung cancer progression, underscoring its potential as an innovative therapeutic target and diagnostic biomarker. Targeting MALAT1 alone or combined with existing therapies holds promise to mitigate lung cancer progression and improve patient outcomes.
Collapse
Affiliation(s)
- Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur 302017, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India
| | - Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur 302017, India
| | - Haider Ali
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Shakir Saleem
- Department of Public Health. College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Vijaya Paul Samuel
- Department of Anatomy, RAK College of Medicine, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE
| | | | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar Sunway, Selangor Darul Ehsan 47500, Malaysia
| |
Collapse
|
7
|
Zhao S, Li S, Yang J, Gao W, Chen Z. GM-CSF-mediated inducement of bone marrow MDSCs by TSA and effect on survival of graft in mice. Eur J Med Res 2022; 27:161. [PMID: 36031660 PMCID: PMC9422167 DOI: 10.1186/s40001-022-00788-8] [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: 03/02/2022] [Accepted: 08/12/2022] [Indexed: 11/10/2022] Open
Abstract
Objective This study analyzed the effect of HDAC inhibitor, trichostatin A (TSA), in inducing granulocyte–macrophage colony-stimulating factor (GM-CSF)-mediated bone marrow (BM) cell differentiation to myeloid-derived suppressor cells (MDSCs) in vitro and in vivo. Methods BM cell differentiation to CD11b + GR-1 + MDSCs was achieved by in vitro culture with TSA and GM-CSF, and the collected cells were analyzed by mixed lymphocyte culture to identify suppressive actions against effector T cells. RT-PCR and ELISA were conducted to analyze the CCL5 mRNA and protein levels in TSA + GM-CSF + BM, GR-1 + MDSCs and GR-1 + MDSC + CCL5 groups. The survival of cardiac grafts was compared between groups. Results TSA was beneficial for the GM-CSF-mediated BM differentiation to CD11b + GR-1 + MDSCs. Adoptive transfer of GR-1 + MDSCs was powerful in suppressing CD4 + CD25-T cell proliferation and the effect was mediated by iNOS and HO-1; it also increased CCL5 gradient concentration between grafts and plasma to recruit Treg to grafts and prolong the survival of the grafts. Survival analysis revealed that the survival of grafts after adoptive transfer of GR-1 + MDSCs could be prolonged. Conclusion This study helps in further research on the application value of MDSCs in the field of transplant, and may provide a new thought for the cell therapy in inducing immune tolerance in organ transplant.
Collapse
Affiliation(s)
- Shuguang Zhao
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China.
| | - Shaohua Li
- Department of Respiratory Medicine, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingci Yang
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Weinian Gao
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| | - Ziying Chen
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, China
| |
Collapse
|
8
|
Luo W, Napoleon JV, Zhang F, Lee YG, Wang B, Putt KS, Low PS. Repolarization of Tumor-Infiltrating Myeloid Cells for Augmentation of CAR T Cell Therapies. Front Immunol 2022; 13:816761. [PMID: 35250995 PMCID: PMC8889096 DOI: 10.3389/fimmu.2022.816761] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/20/2022] [Indexed: 12/21/2022] Open
Abstract
Although CAR T cell therapies have proven to be effective in treating hematopoietic cancers, their abilities to regress solid tumors have been less encouraging. Mechanisms to explain these disparities have focused primarily on differences in cancer cell heterogeneity, barriers to CAR T cell penetration of solid tumors, and immunosuppressive microenvironments. To evaluate the contributions of immunosuppressive tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) on CAR T cell efficacies, we have exploited the ability of a folate-targeted Toll-like receptor 7 agonist (FA-TLR7-1A) to specifically reactivate TAMs and MDSCs from an immunosuppressive to pro-inflammatory phenotype without altering the properties of other immune cells. We report here that FA-TLR7-1A significantly augments standard CAR T cell therapies of 4T1 solid tumors in immune competent mice. We further show that co-administration of the FA-TLR7-1A with the CAR T cell therapy not only repolarizes TAMs and MDSCs from an M2-like anti-inflammatory to M1-like pro-inflammatory phenotype, but also enhances both CAR T cell and endogenous T cell accumulation in solid tumors while concurrently increasing their states of activation. Because analogous myeloid cells in healthy tissues ar not altered by administration of FA-TLR7-1A, no systemic activation of the immune system nor accompanying weight loss is observed. These data argue that immunosuppressive myeloid cells contribute prominently to the failure of CAR T cells to eradicate solid tumors and suggest that methods to reprogram tumor associated myeloid cells to a more inflammatory phenotype could significantly augment the potencies of CAR T cell therapies.
Collapse
Affiliation(s)
- Weichuan Luo
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - John V Napoleon
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Fenghua Zhang
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Yong Gu Lee
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Bingbing Wang
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Karson S Putt
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Philip S Low
- Department of Chemistry, Purdue Institute for Drug Discovery and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
9
|
Iglesias-Escudero M, Segundo DS, Merino-Fernandez D, Mora-Cuesta VM, Lamadrid P, Alonso-Peña M, Raso S, Iturbe D, Fernandez-Rozas S, Cifrian J, López-Hoyos M. Myeloid-Derived Suppressor Cells Are Increased in Lung Transplant Recipients and Regulated by Immunosuppressive Therapy. Front Immunol 2022; 12:788851. [PMID: 35185863 PMCID: PMC8848105 DOI: 10.3389/fimmu.2021.788851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/17/2021] [Indexed: 12/28/2022] Open
Abstract
Lung transplantation remains as a primary treatment for end-stage lung diseases. Although remarkable improvement has been achieved due to the immunosuppressive protocols, long-term survival for lung transplant recipients (LTR) is still limited. In the last few decades, an increasing interest has grown in the study of dysregulation of immune mechanisms underlying allograft failure. In this regard, myeloid-derived suppressor cells (MDSCs) could play an important role in the promotion of graft tolerance due to their immune regulatory function. Here, we describe for the first time circulating subsets MDSCs from LTR at several time points and we evaluate the relationship of MDSCs with sort-term lung transplant outcomes. Although no effect of MDSCs subsets on short-term clinical events was observed, our results determine that Mo-MDSCs frequencies are increased after acute cellular rejection (ACR), suggesting a possible role for Mo-MDSCs in the development of chronic lung allograft dysfunction (CLAD). Therefore, whether MDSCs subsets play a role as biomarkers of chronic rejection remains unknown and requires further investigations. Also, the effects of the different immunosuppressive treatments on these subpopulations remain under research and further studies are needed to establish to what extend MDSCs immune modulation could be responsible for allograft acceptance.
Collapse
Affiliation(s)
- María Iglesias-Escudero
- Transplant and Autoimmunity group, Research Institute-IDIVAL, Santander, Spain.,Immunology Department, Universitary Hospital Germans Trias i Pujol, Badalona, Spain
| | - David San Segundo
- Transplant and Autoimmunity group, Research Institute-IDIVAL, Santander, Spain.,Immunology Department, Universitary Hospital Marqués de Valdecilla-IDIVAL, Santander, Spain
| | | | - Victor M Mora-Cuesta
- Pneumology Department, Universitary Hospital Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Patricia Lamadrid
- Transplant and Autoimmunity group, Research Institute-IDIVAL, Santander, Spain
| | - Marta Alonso-Peña
- Transplant and Autoimmunity group, Research Institute-IDIVAL, Santander, Spain
| | - Sandra Raso
- Transplant and Autoimmunity group, Research Institute-IDIVAL, Santander, Spain
| | - David Iturbe
- Pneumology Department, Universitary Hospital Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Sonia Fernandez-Rozas
- Pneumology Department, Universitary Hospital Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Jose Cifrian
- Pneumology Department, Universitary Hospital Marqués de Valdecilla-IDIVAL, Santander, Spain
| | - Marcos López-Hoyos
- Transplant and Autoimmunity group, Research Institute-IDIVAL, Santander, Spain.,Immunology Department, Universitary Hospital Marqués de Valdecilla-IDIVAL, Santander, Spain.,Molecular Biology Department, Universidad Cantabria, Santander, Spain
| |
Collapse
|
10
|
Bhat DK, Olkhanud PB, Gangaplara A, Seifuddin F, Pirooznia M, Biancotto A, Fantoni G, Pittman C, Francis B, Dagur PK, Saxena A, McCoy JP, Pfeiffer RM, Fitzhugh CD. Early Myeloid Derived Suppressor Cells (eMDSCs) Are Associated With High Donor Myeloid Chimerism Following Haploidentical HSCT for Sickle Cell Disease. Front Immunol 2021; 12:757279. [PMID: 34917079 PMCID: PMC8669726 DOI: 10.3389/fimmu.2021.757279] [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: 08/11/2021] [Accepted: 11/11/2021] [Indexed: 12/24/2022] Open
Abstract
Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) is a widely available curative option for patients with sickle cell disease (SCD). Our original non-myeloablative haplo-HSCT trial employing post-transplant (PT) cyclophosphamide had a low incidence of GVHD but had high rejection rates. Here, we aimed to evaluate immune reconstitution following haplo-HSCT and identify cytokines and cells associated with graft rejection/engraftment. 50 cytokines and 10 immune cell subsets were screened using multiplex-ELISA and flow cytometry, respectively, at baseline and PT-Days 30, 60, 100, and 180. We observed the most significant differences in cytokine levels between the engrafted and rejected groups at PT-Day 60, corresponding with clinical findings of secondary graft rejection. Of the 44 cytokines evaluated, plasma concentrations of 19 cytokines were different between the two groups at PT-Day 60. Factor analysis suggested two independent factors. The first factor (IL-17A, IL-10, IL-7, G-CSF, IL-2, MIP-1a, VEGF, and TGFb1 contributed significantly) was strongly associated with engraftment with OR = 2.7 (95%CI of 1.4 to 5.4), whereas the second factor (GROa and IL-18 contributed significantly) was not significantly associated with engraftment. Sufficient donor myeloid chimerism (DMC) is critical for the success of HSCT; here, we evaluated immune cells among high (H) DMC (DMC≥20%) and low (L) DMC (DMC<20%) groups along with engrafted and rejected groups. We found that early myeloid-derived suppressor cell (eMDSC) frequencies were elevated in engrafted patients and patients with HDMC at PT-Day 30 (P< 0.04 & P< 0.003, respectively). 9 of 20 patients were evaluated for the source of eMDSCs. The HDMC group had high mixed chimeric eMDSCs as compared to the LDMC group (P< 0.00001). We found a positive correlation between the frequencies of eMDSCs and Tregs at PT-Day 100 (r=0.72, P <0.0007); eMDSCs at BSL and Tregs at PT-Day 100 (r=0.63, P <0.004). Of 10 immune regulatory cells and 50 cytokines, we observed mixed chimeric eMDSCs and IL-17A, IL-10, IL-7, G-CSF, IL-2, MIP-1a, VEGF, TGFb1 as potential hits which could serve as prognostic markers in predicting allograft outcome towards engraftment following haploidentical HSCT employing post-transplant cyclophosphamide. The current findings need to be replicated and further explored in a larger cohort.
Collapse
Affiliation(s)
- Deepali K Bhat
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Purevdorj B Olkhanud
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Arunakumar Gangaplara
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Fayaz Seifuddin
- Bioinformatics and Computational Biology Core Facility, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Mehdi Pirooznia
- Bioinformatics and Computational Biology Core Facility, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Angélique Biancotto
- Center for Human Immunology, Autoimmunity, and Inflammation, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Giovanna Fantoni
- Center for Human Immunology, Autoimmunity, and Inflammation, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Corinne Pittman
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Berline Francis
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Pradeep K Dagur
- Flow Cytometry Core, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda MD, United States
| | - Ankit Saxena
- Flow Cytometry Core, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda MD, United States
| | - J Philip McCoy
- Flow Cytometry Core, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda MD, United States
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Courtney D Fitzhugh
- Cellular and Molecular Therapeutics Branch, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
11
|
Li X, Li Y, Yu Q, Qian P, Huang H, Lin Y. Metabolic reprogramming of myeloid-derived suppressor cells: An innovative approach confronting challenges. J Leukoc Biol 2021; 110:257-270. [PMID: 34075637 PMCID: PMC8361984 DOI: 10.1002/jlb.1mr0421-597rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023] Open
Abstract
Immune cells such as T cells, macrophages, dendritic cells, and other immunoregulatory cells undergo metabolic reprogramming in cancer and inflammation-derived microenvironment to meet specific physiologic and functional demands. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that are characterized by immunosuppressive activity, which plays a key role in host immune homeostasis. In this review, we have discussed the core metabolic pathways, including glycolysis, lipid and fatty acid biosynthesis, and amino acid metabolism in the MDSCs under various pathologic situations. Metabolic reprogramming is a determinant of the phenotype and functions of MDSCs, and is therefore a novel therapeutic possibility in various diseases.
Collapse
Affiliation(s)
- Xiaoqing Li
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouZhejiangChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouZhejiangChina
| | - Yixue Li
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouZhejiangChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouZhejiangChina
| | - Qinru Yu
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouZhejiangChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouZhejiangChina
| | - Pengxu Qian
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouZhejiangChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouZhejiangChina
| | - He Huang
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouZhejiangChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouZhejiangChina
| | - Yu Lin
- Bone Marrow Transplantation Center, the First Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouZhejiangChina
- Institute of HematologyZhejiang UniversityHangzhouZhejiangChina
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity TherapyHangzhouZhejiangChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouZhejiangChina
| |
Collapse
|
12
|
Kimm MA, Klenk C, Alunni-Fabbroni M, Kästle S, Stechele M, Ricke J, Eisenblätter M, Wildgruber M. Tumor-Associated Macrophages-Implications for Molecular Oncology and Imaging. Biomedicines 2021; 9:biomedicines9040374. [PMID: 33918295 PMCID: PMC8066018 DOI: 10.3390/biomedicines9040374] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor-associated macrophages (TAMs) represent the largest group of leukocytes within the tumor microenvironment (TME) of solid tumors and orchestrate the composition of anti- as well as pro-tumorigenic factors. This makes TAMs an excellent target for novel cancer therapies. The plasticity of TAMs resulting in varying membrane receptors and expression of intracellular proteins allow the specific characterization of different subsets of TAMs. Those markers similarly allow tracking of TAMs by different means of molecular imaging. This review aims to provides an overview of the origin of tumor-associated macrophages, their polarization in different subtypes, and how characteristic markers of the subtypes can be used as targets for molecular imaging and theranostic approaches.
Collapse
Affiliation(s)
- Melanie A. Kimm
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Christopher Klenk
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Marianna Alunni-Fabbroni
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Sophia Kästle
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Matthias Stechele
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
| | - Michel Eisenblätter
- Department of Diagnostic and Interventional Radiology, Freiburg University Hospital, 79106 Freiburg, Germany;
| | - Moritz Wildgruber
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.A.K.); (C.K.); (M.A.-F.); (S.K.); (M.S.); (J.R.)
- Correspondence: ; Tel.: +49-0-89-4400-76640
| |
Collapse
|
13
|
Han D, Tao J, Fu R, Shao Z. Myeloid-derived suppressor cell cytokine secretion as prognostic factor in myelodysplastic syndromes. Innate Immun 2020; 26:703-715. [PMID: 33050756 PMCID: PMC7787555 DOI: 10.1177/1753425920961157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that play a critical immunosuppressive role in the tumour micro-environment. Although biological research on MDSCs has made progress, the relationship between the secretion of cytokines by MDSCs and poor prognosis is not clear, and there are no criteria to measure the functional status of MDSCs. Here, we detected the mRNA expression of IL-10, IL-12, TGF-β and TNF-α in MDSCs and the levels of these cytokines in MDSC culture supernatants of patients with myelodysplastic syndromes, and quantified the functional status of MDSCs by IL-10/IL-12 ratio and TGF-β/TNF-α ratio. We found that the ratio of IL-10/IL-12 and TGF-β/TNF-α was significantly higher in higher-risk MDS than in lower-risk MDS and normal control groups. The TGF-β/TNF-α ratio in MDSCs was positively correlated with the percentage of blast cells and was negatively correlated with the percentage of CD3+CD8+ T lymphocytes. Meanwhile, the TGF-β/TNF-α ratio was higher in patients with a lower absolute neutrophil count. It suggested that MDSCs in higher-risk MDS have a stronger immunosuppressive effect and might be related to poor prognosis. Quantifying the functional status of MDSCs with IL-10/IL-12 and TGF-β/TNF-α ratio might help to evaluate the balance of cellular immunity of MDSCs in MDS.
Collapse
Affiliation(s)
- Dong Han
- Department of Hematology, Tianjin Medical University General Hospital, PR China.,Department of Lymphoma, Beijing Shijitan Hospital, Capital Medical University, PR China
| | - Jinglian Tao
- Department of Hematology, Tianjin Medical University General Hospital, PR China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, PR China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, PR China
| |
Collapse
|
14
|
Davidov V, Jensen G, Mai S, Chen SH, Pan PY. Analyzing One Cell at a TIME: Analysis of Myeloid Cell Contributions in the Tumor Immune Microenvironment. Front Immunol 2020; 11:1842. [PMID: 32983100 PMCID: PMC7492293 DOI: 10.3389/fimmu.2020.01842] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/09/2020] [Indexed: 12/30/2022] Open
Abstract
Tumor-mediated regulation of the host immune system involves an intricate signaling network that results in the tumor's inherent survival benefit. Myeloid cells are central in orchestrating the mechanisms by which tumors escape immune detection and continue their proliferative programming. Myeloid cell activation has historically been classified using a dichotomous system of classical (M1-like) and alternative (M2-like) states, defining general pro- and anti-inflammatory functions, respectively. Explosions in bioinformatics analyses have rapidly expanded the definitions of myeloid cell pro- and anti-inflammatory states with different combinations of tissue- and disease-specific phenotypic and functional markers. These new definitions have allowed researchers to target specific subsets of disease-propagating myeloid cells in order to modify or arrest the natural progression of the associated disease, especially in the context of tumor-immune interactions. Here, we discuss the myeloid cell contribution to solid tumor initiation and maintenance, and strategies to reprogram their phenotypic and functional fate, thereby disabling the network that benefits tumor survival.
Collapse
Affiliation(s)
- Vitaliy Davidov
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Garrett Jensen
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Sunny Mai
- Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Shu-Hsia Chen
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| | - Ping-Ying Pan
- Texas A&M College of Medicine, Bryan, TX, United States.,Center for Immunotherapy Research, Cancer Center of Excellence, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
15
|
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: 3.4] [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
|
16
|
Sinha A, Singh V, Tandon R, Mohan Srivastava L. Dichotomy of complement system: Tumorigenesis or destruction. Immunol Lett 2020; 223:89-96. [PMID: 32333965 DOI: 10.1016/j.imlet.2020.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 04/06/2020] [Accepted: 04/18/2020] [Indexed: 01/12/2023]
Abstract
Complement system proteins, their regulators and endpoint effector complex significantly promote tumor growth by upregulation of oncogenic growth factors, activation of mitogenic signalling pathways and breakage of normal cell cycle. Contrastingly, complement cascades, initiated by anti-tumor therapeutic antibodies, also play a pivotal role in therapy response. This contradictory role of complement system possibly be a very crucial factor for the outcomes of antibody mediated immunotherapies. Herein, we reviewed the twin role of the complement system in cancer and also the genetic variations in complement system genes. Future studies should be focused on the biomarker discovery for the personalised cancer immunotherapies.
Collapse
Affiliation(s)
- Ashima Sinha
- Department of BiochemIstry, Sir Ganga Ram Hospital, New Delhi-110060, India; SAGE Publications India Pvt Ltd., New Delhi-110044, India
| | - Virendra Singh
- Laboratory of Precision Medicine Research, School of Biotechnology, Jawaharlal Nehru University, New Delhi-110067, India.
| | - Ravi Tandon
- Laboratory of AIDS research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi-110067, India
| | - Lalit Mohan Srivastava
- Department of Biochemistry and Lab Medicine, Sir Ganga Ram Kolmet Hospital, New Delhi-110005, India.
| |
Collapse
|
17
|
Yan JJ, Ryu JH, Piao H, Hwang JH, Han D, Lee SK, Jang JY, Lee J, Koo TY, Yang J. Granulocyte Colony-Stimulating Factor Attenuates Renal Ischemia-Reperfusion Injury by Inducing Myeloid-Derived Suppressor Cells. J Am Soc Nephrol 2020; 31:731-746. [PMID: 32132198 DOI: 10.1681/asn.2019060601] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/22/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Granulocyte colony-stimulating factor (G-CSF) can increase populations of myeloid-derived suppressor cells, innate immune suppressors that play an immunoregulatory role in antitumor immunity. However, the roles of myeloid-derived suppressor cells and G-CSF in renal ischemia-reperfusion injury remain unclear. METHODS We used mouse models of ischemia-reperfusion injury to investigate whether G-CSF can attenuate renal injury by increasing infiltration of myeloid-derived suppressor cells into kidney tissue. RESULTS G-CSF treatment before ischemia-reperfusion injury subsequently attenuated acute renal dysfunction, tissue injury, and tubular apoptosis. Additionally, G-CSF treatment suppressed renal infiltration of macrophages and T cells as well as renal levels of IL-6, MCP-1, IL-12, TNF-α, and IFN-γ, but it increased levels of IL-10, arginase-1, and reactive oxygen species. Moreover, administering G-CSF after ischemia-reperfusion injury improved the recovery of renal function and attenuated renal fibrosis on day 28. G-CSF treatment increased renal infiltration of myeloid-derived suppressor cells (F4/80-CD11b+Gr-1int), especially the granulocytic myeloid-derived suppressor cell population (CD11b+Ly6GintLy6Clow); splenic F4/80-CD11b+Gr-1+ cells sorted from G-CSF-treated mice displayed higher levels of arginase-1, IL-10, and reactive oxygen species relative to those from control mice. Furthermore, these splenic cells effectively suppressed in vitro T cell activation mainly through arginase-1 and reactive oxygen species, and their adoptive transfer attenuated renal injury. Combined treatment with anti-Gr-1 and G-CSF showed better renoprotective effects than G-CSF alone, whereas preferential depletion of myeloid-derived suppressor cells by pep-G3 or gemcitabine abrogated the beneficial effects of G-CSF against renal injury. CONCLUSIONS G-CSF induced renal myeloid-derived suppressor cells, thereby attenuating acute renal injury and chronic renal fibrosis after ischemia-reperfusion injury. These results suggest therapeutic potential of myeloid-derived suppressor cells and G-CSF in renal ischemia-reperfusion injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Joongyub Lee
- Department of Prevention and Management, Inha University Hospital School of Medicine, Inha University, Incheon, South Korea; and
| | - Tai Yeon Koo
- Biomedical Research Institute and.,Transplantation Center and
| | - Jaeseok Yang
- Biomedical Research Institute and .,Transplantation Center and.,Department of Surgery, Seoul National University Hospital, Seoul, South Korea.,Transplantation Research Institute, Seoul National University College of Medicine, Republic of, South Korea
| |
Collapse
|
18
|
Emerging Role of Myeloid-derived Suppressor Cells in the Biology of Transplantation Tolerance. Transplantation 2020; 104:467-475. [DOI: 10.1097/tp.0000000000002996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
19
|
Suk Lee Y, Davila E, Zhang T, Milmoe HP, Vogel SN, Bromberg JS, Scalea JR. Myeloid-derived suppressor cells are bound and inhibited by anti-thymocyte globulin. Innate Immun 2019; 25:46-59. [PMID: 30782043 PMCID: PMC6830891 DOI: 10.1177/1753425918820427] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) inhibit T cell responses and are
relevant to cancer, autoimmunity and transplant biology. Anti-thymocyte globulin
(ATG) is a commonly used T cell depletion agent, yet the effect of ATG on MDSCs
has not been investigated. MDSCs were generated in Lewis Lung Carcinoma 1
tumor-bearing mice. MDSC development and function were assessed in
vivo and in vitro with and without ATG
administration. T cell suppression assays, RT-PCR, flow cytometry and arginase
activity assays were used to assess MDSC phenotype and function. MDSCs increased
dramatically in tumor-bearing mice and the majority of splenic MDSCs were of the
polymorphonuclear subset. MDSCs potently suppressed T cell proliferation.
ATG-treated mice developed 50% fewer MDSCs and these MDSCs were significantly
less suppressive of T cell proliferation. In vitro, ATG
directly bound 99.6% of MDSCs. CCR7, L-selectin and LFA-1 were expressed by both
T cells and MDSCs, and binding of LFA-1 was inhibited by ATG pre-treatment.
Arg-1 and PD-L1 transcript expression were reduced 30–40% and arginase activity
decreased in ATG-pretreated MDSCs. MDSCs were bound and functionally inhibited
by ATG. T cells and MDSCs expressed common Ags which were also targets of ATG.
ATG may be helpful in tumor models seeking to suppress MDSCs. Alternatively, ATG
may inadvertently inhibit important T cell regulatory events in autoimmunity and
transplantation.
Collapse
Affiliation(s)
- Young Suk Lee
- 1 Department of Surgery, University of Maryland, Baltimore, USA
| | - Eduardo Davila
- 2 Department of Microbiology and Immunology, University of Maryland, Baltimore, USA
| | - Tianshu Zhang
- 1 Department of Surgery, University of Maryland, Baltimore, USA
| | - Hugh P Milmoe
- 1 Department of Surgery, University of Maryland, Baltimore, USA
| | - Stefanie N Vogel
- 2 Department of Microbiology and Immunology, University of Maryland, Baltimore, USA
| | - Jonathan S Bromberg
- 1 Department of Surgery, University of Maryland, Baltimore, USA.,2 Department of Microbiology and Immunology, University of Maryland, Baltimore, USA
| | - Joseph R Scalea
- 1 Department of Surgery, University of Maryland, Baltimore, USA.,2 Department of Microbiology and Immunology, University of Maryland, Baltimore, USA
| |
Collapse
|
20
|
Cao L, Dai C, Qin R, Guo Y, Liu J. (3R)-5,6,7-trihydroxy-3-isopropyl-3-methylisochroman-1-one enhanced the therapeutic efficacy of anti-PD1 antibody through inhibiting PI3Kδ/γ. Immunopharmacol Immunotoxicol 2019; 41:599-606. [PMID: 31691624 DOI: 10.1080/08923973.2019.1678634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purpose: Immunotherapy has demonstrated durable clinical responses in various cancers by disinhibiting the immune system, largely attributed to the success of immune-checkpoint blockade. However, there are still subsets of patients across multiple cancers not showing robust responses to these agents and one significant barrier to their efficacy may be the recruitment of myeloid-derived suppressor cells (MDSCs) into the tumor microenvironment. In this study, we demonstrated that functional inhibition of MDSCs with (3 R)-5,6,7-trihydroxy-3-isopropyl-3-methylisochroman-1-one (TIMO), a potent PI3Kδ/γ inhibitor, enhanced the therapeutic efficacy of anti-PD1 antibody in the tumor model.Materials and methods: A syngeneic ovarian tumor model was established. MDSCs from the peripheral blood and tumor parenchyma were analyzed by flow cytometry. Proliferation and killing effects of T-lymphocytes were measured. IFNγ production was measured by ELISA assay. qPCR and western blot were used to detect the gene and protein expression. Furthermore, the therapeutic effects of TIMO combined with anti-PD1 antibody were assessed by the tumor model.Results: Our data demonstrated that inhibition of granulocytic myeloid-derived suppressor cells (G-MDSCs) function with TIMO could overcome MDSCs-mediated immunosuppression and promote antigen-specific T-lymphocyte responses, resulting in the restoration of cytotoxic T cell-mediated tumor control. We further demonstrated that TIMO and anti-PD1 combination therapy promoted tumor growth control in a syngeneic ovarian tumor model.Conclusions: Our results provided proof of concept for a new combination strategy involving the use of a selective inhibitor of PI3Kδ/γ to inhibit the function of MDSCs to enhance tumor responses to immune checkpoint blocking antibodies.
Collapse
Affiliation(s)
- Lu Cao
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - ChunMei Dai
- Department of School Hospital, Changchun University of Chinese Medicine, Changchun, China
| | - Rui Qin
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - YaHua Guo
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - JunBao Liu
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Goulart MR, Hlavaty SI, Chang YM, Polton G, Stell A, Perry J, Wu Y, Sharma E, Broxholme J, Lee AC, Szladovits B, Turmaine M, Gribben J, Xia D, Garden OA. Phenotypic and transcriptomic characterization of canine myeloid-derived suppressor cells. Sci Rep 2019; 9:3574. [PMID: 30837603 PMCID: PMC6400936 DOI: 10.1038/s41598-019-40285-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 02/13/2019] [Indexed: 01/19/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are key players in immune evasion, tumor progression and metastasis. MDSCs accumulate under various pathological states and fall into two functionally and phenotypically distinct subsets that have been identified in humans and mice: polymorphonuclear (PMN)-MDSCs and monocytic (M)-MDSCs. As dogs are an excellent model for human tumor development and progression, we set out to identify PMN-MDSCs and M-MDSCs in clinical canine oncology patients. Canine hypodense MHC class II-CD5-CD21-CD11b+ cells can be subdivided into polymorphonuclear (CADO48A+CD14-) and monocytic (CADO48A-CD14+) MDSC subsets. The transcriptomic signatures of PMN-MDSCs and M-MDSCs are distinct, and moreover reveal a statistically significant similarity between canine and previously published human PMN-MDSC gene expression patterns. As in humans, peripheral blood frequencies of canine PMN-MDSCs and M-MDSCs are significantly higher in dogs with cancer compared to healthy control dogs (PMN-MDSCs: p < 0.001; M-MDSCs: p < 0.01). By leveraging the power of evolution, we also identified additional conserved genes in PMN-MDSCs of multiple species that may play a role in MDSC function. Our findings therefore validate the dog as a model for studying MDSCs in the context of cancer.
Collapse
Affiliation(s)
- Michelle R Goulart
- Royal Veterinary College, London, UK
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Sabina I Hlavaty
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - James Perry
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ying Wu
- Royal Veterinary College, London, UK
| | - Eshita Sharma
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - John Broxholme
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Avery C Lee
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Mark Turmaine
- Division of Bioscience, University College London, London, UK
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Dong Xia
- Royal Veterinary College, London, UK
| | - Oliver A Garden
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Llaudo I, Fribourg M, Medof ME, Conde P, Ochando J, Heeger PS. C5aR1 regulates migration of suppressive myeloid cells required for costimulatory blockade-induced murine allograft survival. Am J Transplant 2019; 19:633-645. [PMID: 30106232 PMCID: PMC6375810 DOI: 10.1111/ajt.15072] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/17/2018] [Accepted: 07/31/2018] [Indexed: 01/25/2023]
Abstract
Costimulatory blockade-induced murine cardiac allograft survival requires intragraft accumulation of CD11b+ Ly6Clo Ly6G- regulatory myeloid cells (Mregs) that expand regulatory T cells (Tregs) and suppress effector T cells (Teffs). We previously showed that C5a receptor (C5aR1) signaling on T cells activates Teffs and inhibits Tregs, but whether and/or how C5aR1 affects Mregs required for transplant survival is unknown. Although BALB/c hearts survived >60 days in anti-CD154 (MR1)-treated or cytotoxic T-lymphocyte associated protein 4 (CTLA4)-Ig-treated wild-type (WT) recipients, they were rejected at ~30 days in MR1-treated or CTLA4-Ig-treated recipients selectively deficient in C5aR1 restricted to myeloid cells (C5ar1fl/fl xLysM-Cre). This accelerated rejection was associated with ~2-fold more donor-reactive T cells and ~40% less expansion of donor-reactive Tregs. Analysis of graft-infiltrating mononuclear cells on posttransplant day 6 revealed fewer Ly6Clo monocytes in C5ar1fl/fl xLysM-Cre recipients. Expression profiling of intragraft Ly6Clo monocytes showed that C5aR1 deficiency downregulated genes related to migration/locomotion without changes in genes associated with suppressive function. Cotransfer of C5ar1fl/fl and C5ar1fl/fl xLysM-Cre myeloid cells into MR1-treated allograft recipients resulted in less accumulation of C5ar1-/- cells within the allografts, and in vitro assays confirmed that Ly6Chi myeloid cells migrate to C5a/C5aR1-initiated signals. Together, our results newly link myeloid cell-expressed C5aR1 to intragraft accumulation of myeloid cells required for prolongation of heart transplant survival induced by costimulatory blockade.
Collapse
Affiliation(s)
- Ines Llaudo
- Translational Transplant Research Center,,Department of Medicine, and Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY
| | - Miguel Fribourg
- Translational Transplant Research Center,,Department of Neurology, Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY
| | - M. Edward Medof
- Institute of Pathology, Case Western Reserve University, Cleveland OH
| | - Patricia Conde
- Department of Medicine, and Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jordi Ochando
- Department of Medicine, and Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY
| | - Peter S. Heeger
- Translational Transplant Research Center,,Department of Medicine, and Immunology Institute Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
23
|
Li M, Zhu D, Wang T, Xia X, Tian J, Wang S. Roles of Myeloid-Derived Suppressor Cell Subpopulations in Autoimmune Arthritis. Front Immunol 2018; 9:2849. [PMID: 30564242 PMCID: PMC6288996 DOI: 10.3389/fimmu.2018.02849] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/19/2018] [Indexed: 12/29/2022] Open
Abstract
Emerging evidence suggests the promise of the use of myeloid-derived suppressor cells (MDSCs) in inflammatory disorders based on their unique immune-intervention properties. However, the roles of MDSCs in autoimmune arthritis are not completely understood. Indeed, their immunosuppressive functions in arthritic conditions remain controversial, with heterogeneity among MDSCs and differential effects among subpopulations receiving much attention. As a result, it is necessary to determine the roles of MDSC subpopulations in autoimmune arthritis to clarify their diagnostic and therapeutic potential. Interestingly, in the inflammation niche of autoimmune arthritis, each MDSC subpopulation can exhibit both alternatives of a given characteristic. Moreover, polymorphonuclear MDSCs (PMN-MDSCs) are likely to be more suppressive and stable compared with monocytic MDSCs (MO-MDSCs). Although various important cytokines associated with the differentiation of MDSCs or MDSC subpopulations from immature myeloid precursors, such as granulocyte colony-stimulating factor (G-CSF), have been largely applied in external inductive systems, their roles are not entirely clear. Moreover, MDSC-based clinical treatments in rheumatoid arthritis (RA) continue to represent a significant challenge, as also reported for other autoimmune diseases. In this review, we describe the effects and actions of MDSC subpopulations on the development of autoimmune arthritis and analyze several types of MDSC-based therapeutic strategies to provide comprehensive information regarding immune networks and a foundation for more effective protocols for autoimmune arthritis.
Collapse
Affiliation(s)
- Min Li
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Dongwei Zhu
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Tingting Wang
- Department of Laboratory Medicine, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Children's Hospital, Wuxi, China
| | - Xueli Xia
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jie Tian
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
24
|
Lee HJ, Park SY, Jeong HJ, Kim HJ, Kim MK, Oh JY. Glucocorticoids induce corneal allograft tolerance through expansion of monocytic myeloid-derived suppressor cells. Am J Transplant 2018; 18:3029-3037. [PMID: 30019411 DOI: 10.1111/ajt.15026] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/24/2018] [Accepted: 07/10/2018] [Indexed: 01/25/2023]
Abstract
Glucocorticoids (GCs) are the most widely used drugs to prevent transplant rejection; however, it is not yet clear how GCs induce immune tolerance in transplantation. Here, we demonstrate that GCs induce tolerance to corneal allografts in mice through expansion of MHC class II- CD11b+ Ly6C+ monocytes in the bone marrow and mobilization of the cells to spleen, draining lymph nodes, and graft site. The GC-induced CD11b+ Ly6C+ monocytes inhibited T cell proliferation in vitro, and adoptive transfer of the cells improved the survival of corneal allografts. Depletion of CD11b+ Ly6C+ cells in mice during GC treatment abrogated the effects of GCs in prevention of immune rejection. Together, the results identify monocytic myeloid-derived suppressor cells as crucial mediators of the GC-induced tolerance in transplantation.
Collapse
Affiliation(s)
- Hyun Ju Lee
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea
| | - Se Yeon Park
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea
| | - Hyun Jeong Jeong
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea
| | - Hyeon Ji Kim
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea
| | - Mee Kum Kim
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea.,Department of Ophthalmology, Seoul National University Hospital, Seoul, Korea
| | - Joo Youn Oh
- Laboratory of Ocular Regenerative Medicine and Immunology, Seoul Artificial Eye Center, Seoul National University Hospital Biomedical Research Institute, Seoul, Korea.,Department of Ophthalmology, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
25
|
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: 6.7] [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
|
26
|
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immunosuppressive cells of the myeloid lineage upregulated by mediators of inflammation, such as IL-2, granulocyte colony-stimulating factor, and S100A8/A9. These cells have been studied extensively by tumor biologists. Because of their robust immunosuppressive potential, MDSCs have stirred recent interest among transplant immunologists as well. MDSCs inhibit T-cell responses through, among other mechanisms, the activity of arginase-1 and inducible nitric oxide synthase, and the expansion of T regulatory cells. In the context of transplantation, MDSCs have been studied in several animal models, and to a lesser degree in humans. Here, we will review the immunosuppressive qualities of this important cell type and discuss the relevant studies of MDSCs in transplantation. It may be possible to exploit the immunosuppressive capacity of MDSCs for the benefit of transplant patients.
Collapse
|
27
|
Specific inhibition of PI3Kδ/γ enhances the efficacy of anti-PD1 against osteosarcoma cancer. J Bone Oncol 2018; 16:100206. [PMID: 31334002 PMCID: PMC6617297 DOI: 10.1016/j.jbo.2018.11.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/03/2018] [Accepted: 11/05/2018] [Indexed: 02/06/2023] Open
Abstract
Impressive responses have been observed in patients with cancer treated with checkpoint inhibitory anti-programmed cell death-1 (PD-1) or anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) antibodies through disinhibiting the immune system. However, tumors possess complex immunosuppressive tumor microenvironment to present therapeutic obstacles and the response rates to immune checkpoint inhibition remain low. One significant barrier to the efficacy of anti-PD1 treatment is the recruitment of myeloid-derived suppressor cells (MDSCs) into the tumor. MDSCs dramatically increased in peripheral blood of patients with osteosarcoma and prohibited both T-cell activation and infiltration. Here we demonstrated functional inhibition of G-MDSCs with (S)-(-)-N-[2-(3-Hydroxy-1H-indol-3-yl)-methyl]-acetamide (SNA), a specific inhibitor of PI3Kδ/γ, could prime tumor microenvironment, resultantly enhancing the therapeutic efficacy of anti-PD1 treatment in a syngeneic osteosarcoma tumor model. Combining SNA with anti-PD1 dramatically slowed osteosarcoma tumor growth and prolonged survival time of tumor-bearing mice, at least in part mediated through CD8+ T cells. Our results demonstrated that addition of SNA to anti-PD1 significantly altered infiltration and function of innate immune cells, providing the rationale for combination therapy in patients with osteosarcoma through inhibiting the function of MDSCs with a selective PI3Kδ/γ inhibitor to enhance responses to immune checkpoint blockade.
Collapse
|
28
|
Lin H, Wu Y, Chen J, Huang S, Wang Y. (−)-4-O-(4-O-β-D-glucopyranosylcaffeoyl) Quinic Acid Inhibits the Function of Myeloid-Derived Suppressor Cells to Enhance the Efficacy of Anti-PD1 against Colon Cancer. Pharm Res 2018; 35:183. [DOI: 10.1007/s11095-018-2459-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 07/09/2018] [Indexed: 01/04/2023]
|
29
|
Nguyen A, Ho L, Workenhe ST, Chen L, Samson J, Walsh SR, Pol J, Bramson JL, Wan Y. HDACi Delivery Reprograms Tumor-Infiltrating Myeloid Cells to Eliminate Antigen-Loss Variants. Cell Rep 2018; 24:642-654. [PMID: 30021162 DOI: 10.1016/j.celrep.2018.06.040] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 05/02/2018] [Accepted: 06/08/2018] [Indexed: 01/05/2023] Open
Abstract
Immune recognition of tumor-expressed antigens by cytotoxic CD8+ T cells is the foundation of adoptive T cell therapy (ACT) and has been shown to elicit significant tumor regression. However, therapy-induced selective pressure can sculpt the antigenicity of tumors, resulting in outgrowth of variants that lose the target antigen. We demonstrate that tumor relapse from ACT and subsequent oncolytic viral vaccination can be prevented using class I HDACi, MS-275. Drug delivery subverted the phenotype of tumor-infiltrating CD11b+ Ly6Chi Ly6G- myeloid cells, favoring NOS2/ROS secretion and pro-inflammatory genes characteristic of M1 polarization. Simultaneously, MS-275 abrogated the immunosuppressive function of tumor-infiltrating myeloid cells and reprogrammed them to eliminate antigen-negative tumor cells in a caspase-dependent manner. Elevated IFN-γ within the tumor microenvironment suggests that MS-275 modulates the local cytokine landscape to favor antitumor myeloid polarization through the IFN-γR/STAT1 signaling axis. Exploiting tumor-infiltrating myeloid cell plasticity thus complements T cell therapy in targeting tumor heterogeneity and immune escape.
Collapse
Affiliation(s)
- Andrew Nguyen
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Louisa Ho
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Samuel T Workenhe
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Lan Chen
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | | | - Scott R Walsh
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Jonathan Pol
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Yonghong Wan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
30
|
Negorev D, Beier UH, Zhang T, Quatromoni JG, Bhojnagarwala P, Albelda SM, Singhal S, Eruslanov E, Lohoff FW, Levine MH, Diamond JM, Christie JD, Hancock WW, Akimova T. Human neutrophils can mimic myeloid-derived suppressor cells (PMN-MDSC) and suppress microbead or lectin-induced T cell proliferation through artefactual mechanisms. Sci Rep 2018; 8:3135. [PMID: 29453429 PMCID: PMC5816646 DOI: 10.1038/s41598-018-21450-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/31/2018] [Indexed: 01/01/2023] Open
Abstract
We report that human conventional CD15+ neutrophils can be isolated in the peripheral blood mononuclear cell (PBMC) layer during Ficoll gradient separation, and that they can impair T cell proliferation in vitro without concomitant neutrophil activation and killing. This effect was observed in a total of 92 patients with organ transplants, lung cancer or anxiety/depression, and in 18 healthy donors. Although such features are typically associated in the literature with the presence of certain myeloid-derived suppressor cell (PMN-MDSC) populations, we found that commercial centrifuge tubes that contained membranes or gels for PBMC isolation led to up to 70% PBMC contamination by CD15+ neutrophils, with subsequent suppressive effects in certain cellular assays. In particular, the suppressive activity of human MDSC should not be evaluated using lectin or microbead stimulation, whereas assays involving soluble or plate-bound antibodies or MLR are unaffected. We conclude that CD15+ neutrophil contamination, and associated effects on suppressor assays, can lead to significant artefacts in studies of human PMN-MDSC.
Collapse
Affiliation(s)
- Dmitri Negorev
- The Pathology Bioresource, Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ulf H Beier
- Division of Nephrology, Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania, PA, 19104, Philadelphia, USA
| | - Tianyi Zhang
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Jon G Quatromoni
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Pratik Bhojnagarwala
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Steven M Albelda
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunil Singhal
- Division of Thoracic Surgery, Department of Surgery, University of Pennsylvania, 19104, Philadelphia, PA, USA
| | - Evgeniy Eruslanov
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Falk W Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, 20892-154, USA
| | - Matthew H Levine
- Department of Surgery, Penn Transplant Institute, Hospital of the University of Pennsylvania and University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Joshua M Diamond
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jason D Christie
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wayne W Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA
| | - Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Zhang W, Li J, Qi G, Tu G, Yang C, Xu M. Myeloid-derived suppressor cells in transplantation: the dawn of cell therapy. J Transl Med 2018; 16:19. [PMID: 29378596 PMCID: PMC5789705 DOI: 10.1186/s12967-018-1395-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 01/22/2018] [Indexed: 02/07/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a series of innate cells that play a significant role in inhibiting T cell-related responses. This heterogeneous population of immature cells is involved in tumor immunity. Recently, the function and importance of MDSCs in transplantation have garnered the attention of scientists and have become an important focus of transplantation immunology research because MDSCs play a key role in establishing immune tolerance in transplantation. In this review, we summarize recent studies of MDSCs in different types of transplantation. We also focus on the influence of immunosuppressive drugs on MDSCs as well as future obstacles and research directions in this field.
Collapse
Affiliation(s)
- Weitao Zhang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Jiawei Li
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Guisheng Qi
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Guowei Tu
- Department of Intensive Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Yang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| | - Ming Xu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai Key Laboratory of Organ Transplantation, 180 Fenglin Road, Shanghai, 200032 China
| |
Collapse
|
32
|
Yang L, Wang B, Qin J, Zhou H, Majumdar APN, Peng F. Blockade of CCR5-mediated myeloid derived suppressor cell accumulation enhances anti-PD1 efficacy in gastric cancer. Immunopharmacol Immunotoxicol 2018; 40:91-97. [PMID: 29303012 DOI: 10.1080/08923973.2017.1417997] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE Myeloid derived suppressor cells (MDSC) play an important role in tumor immune evasion and its level significantly increased in patients with gastric cancer. Studies confirmed the associations between MDSC and various cytokines in the peripheral blood. However, little is known about the mechanism drawing MDSC into tumor parenchyma. This study was to analyze the correlation between MDSC subsets and CCR5 level in gastric cancer. MATERIALS AND METHODS G-MDSC and M-MDSC from the peripheral blood and tumor parenchyma were analyzed by flow cytometry. CCR5 ligand CCL5 was detected by ELISA. CCR5 was detected by real-time PCR, western blot and flow cytometry. Furthermore, the therapeutic effects of CCR5 blockade was assessed by the tumor model. RESULTS CCR5 ligand, gene and protein expression of CCR5, and surface expression of CCR5 significantly increased in blood and tumor of tumor-bearing mice, suggesting MDSC may be attracted into the parenchyma by CCL5/CCR5. Anti-CCR5 treatment decreased G-MDSC and M-MDSC in the periphery and tumor. In addition, combination treatment enhanced CD4+ and CD8+ T cell infiltration and decreased the tumor burden of tumor-bearing mice. CONCLUSIONS This study elucidated a possible association between MDSC subsets and CCR5, in addition to provide a new potential target to enhance the efficacy of immunotherapy in patients with gastric cancer.
Collapse
Affiliation(s)
- Liu Yang
- a Department of Gastroenterology, Shanghai Ninth People's Hospital, School of Medicine , Shanghai Jiaotong University , Shanghai , P.R. China
| | - Bing Wang
- b Department of Surgery, Shanghai Ninth People's Hospital, School of Medicine , Shanghai Jiaotong University , Shanghai , P.R. China
| | - Jian Qin
- b Department of Surgery, Shanghai Ninth People's Hospital, School of Medicine , Shanghai Jiaotong University , Shanghai , P.R. China
| | - HengHua Zhou
- c Department of Pathology, Shanghai Ninth People's Hospital, School of Medicine , Shanghai Jiaotong University , Shanghai , P.R. China
| | - Adhip P N Majumdar
- d Division of Gastroenterology and Department of Internal Medicine, Veterans Affairs Medical Center, Karmanos Cancer Institute, School of Medicine , Wayne State University , Detroit , MI , USA
| | - Fei Peng
- e Department of Surgery , Jingan Branch of Huashan Hospital, Fudan University , Shanghai , P.R. China
| |
Collapse
|
33
|
DeBerge M, Zhang S, Glinton K, Grigoryeva L, Hussein I, Vorovich E, Ho K, Luo X, Thorp EB. Efferocytosis and Outside-In Signaling by Cardiac Phagocytes. Links to Repair, Cellular Programming, and Intercellular Crosstalk in Heart. Front Immunol 2017; 8:1428. [PMID: 29163503 PMCID: PMC5671945 DOI: 10.3389/fimmu.2017.01428] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/13/2017] [Indexed: 12/24/2022] Open
Abstract
Phagocytic sensing and engulfment of dying cells and extracellular bodies initiate an intracellular signaling cascade within the phagocyte that can polarize cellular function and promote communication with neighboring non-phagocytes. Accumulating evidence links phagocytic signaling in the heart to cardiac development, adult myocardial homeostasis, and the resolution of cardiac inflammation of infectious, ischemic, and aging-associated etiology. Phagocytic clearance in the heart may be carried out by professional phagocytes, such as macrophages, and non-professional cells, including myofibrolasts and potentially epithelial cells. During cardiac development, phagocytosis initiates growth cues for early cardiac morphogenesis. In diseases of aging, including myocardial infarction, heightened levels of cell death require efficient phagocytic debridement to salvage further loss of terminally differentiated adult cardiomyocytes. Additional risk factors, including insulin resistance and other systemic risk factors, contribute to inefficient phagocytosis, altered phagocytic signaling, and delayed cardiac inflammation resolution. Under such conditions, inflammatory presentation of myocardial antigen may lead to autoimmunity and even possible rejection of transplanted heart allografts. Increased understanding of these basic mechanisms offers therapeutic opportunities.
Collapse
Affiliation(s)
- Matthew DeBerge
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Shuang Zhang
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Kristofor Glinton
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Luba Grigoryeva
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Islam Hussein
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Esther Vorovich
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Karen Ho
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Xunrong Luo
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Edward B Thorp
- Department of Pathology, Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
34
|
Shayan G, Kansy BA, Gibson SP, Srivastava RM, Bryan JK, Bauman JE, Ohr J, Kim S, Duvvuri U, Clump DA, Heron DE, Johnson JT, Hershberg RM, Ferris RL. Phase Ib Study of Immune Biomarker Modulation with Neoadjuvant Cetuximab and TLR8 Stimulation in Head and Neck Cancer to Overcome Suppressive Myeloid Signals. Clin Cancer Res 2017; 24:62-72. [PMID: 29061643 DOI: 10.1158/1078-0432.ccr-17-0357] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/18/2017] [Accepted: 10/17/2017] [Indexed: 01/04/2023]
Abstract
Purpose: The response rate of patients with head and neck squamous cell carcinoma (HNSCC) to cetuximab therapy is only 15% to 20%, despite frequent EGFR overexpression. Because immunosuppression is common in HNSCC, we hypothesized that adding a proinflammatory TLR8 agonist to cetuximab therapy might result in enhanced T-lymphocyte stimulation and anti-EGFR-specific priming.Experimental Design: Fourteen patients with previously untreated HNSCC were enrolled in this neoadjuvant trial and treated preoperatively with 3 to 4 weekly doses of motolimod (2.5 mg/m2) and cetuximab. Correlative tumor and peripheral blood specimens were obtained at baseline and at the time of surgical resection and analyzed for immune biomarker changes. Preclinical in vitro studies were also performed to assess the effect of cetuximab plus motolimod on myeloid cells.Results: TLR8 stimulation skewed monocytes toward an M1 phenotype and reversed myeloid-derived suppressor cell (MDSC) suppression of T-cell proliferation in vitro These data were validated in a prospective phase Ib neoadjuvant trial, in which fewer MDSC and increased M1 monocyte infiltration were found in tumor-infiltrating lymphocytes. Motolimod plus cetuximab also decreased induction of Treg and reduced markers of suppression, including CTLA-4, CD73, and membrane-bound TGFβ. Significantly increased circulating EGFR-specific T cells were observed, concomitant with enhanced CD8+ T-cell infiltration into tumors. These T cells manifested increased T-cell receptor (TCR) clonality, upregulation of the costimulatory receptor CD27, and downregulation of inhibitory receptor TIGIT.Conclusions: Enhanced inflammatory stimulation in the tumor microenvironment using a TLR agonist overcomes suppressive myeloid and regulatory cells, enhancing the cellular antitumor immune response by therapeutic mAb in HNSCC. Clin Cancer Res; 24(1); 62-72. ©2017 AACR.
Collapse
Affiliation(s)
| | - Benjamin A Kansy
- Department of Otolaryngology, Essen University Hospital, Essen, Germany
| | | | | | | | | | - James Ohr
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Seungwon Kim
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania.,Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Umamaheswar Duvvuri
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania.,Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - David A Clump
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
| | | | - Jonas T Johnson
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania.,Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Robert L Ferris
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania. .,Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Wang C, Zhang N, Qi L, Yuan J, Wang K, Wang K, Ma S, Wang H, Lou W, Hu P, Awais M, Cao S, Fu ZF, Cui M. Myeloid-Derived Suppressor Cells Inhibit T Follicular Helper Cell Immune Response in Japanese Encephalitis Virus Infection. THE JOURNAL OF IMMUNOLOGY 2017; 199:3094-3105. [DOI: 10.4049/jimmunol.1700671] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 08/17/2017] [Indexed: 12/23/2022]
|
36
|
Abstract
In addition to being a component of innate immunity and an ancient defense mechanism against invading pathogens, complement activation also participates in the adaptive immune response, inflammation, hemostasis, embryogenesis, and organ repair and development. Activation of the complement system via classical, lectin, or alternative pathways generates anaphylatoxins (C3a and C5a) and membrane attack complex (C5b-9) and opsonizes targeted cells. Complement activation end products and their receptors mediate cell-cell interactions that regulate several biological functions in the extravascular tissue. Signaling of anaphylatoxin receptors or assembly of membrane attack complex promotes cell dedifferentiation, proliferation, and migration in addition to reducing apoptosis. As a result, complement activation in the tumor microenvironment enhances tumor growth and increases metastasis. In this Review, I discuss immune and nonimmune functions of complement proteins and the tumor-promoting effect of complement activation.
Collapse
|
37
|
Effects of Shugan Jianpi Formula (疏肝健脾方) on myeloid-derived suppression cells-mediated depression breast cancer mice. Chin J Integr Med 2016; 23:453-460. [DOI: 10.1007/s11655-016-2734-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Indexed: 11/25/2022]
|
38
|
Wang J, De Veirman K, De Beule N, Maes K, De Bruyne E, Van Valckenborgh E, Vanderkerken K, Menu E. The bone marrow microenvironment enhances multiple myeloma progression by exosome-mediated activation of myeloid-derived suppressor cells. Oncotarget 2016; 6:43992-4004. [PMID: 26556857 PMCID: PMC4791281 DOI: 10.18632/oncotarget.6083] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 10/23/2015] [Indexed: 12/29/2022] Open
Abstract
Exosomes, extracellular nanovesicles secreted by various cell types, modulate the bone marrow (BM) microenvironment by regulating angiogenesis, cytokine release, immune response, inflammation, and metastasis. Interactions between bone marrow stromal cells (BMSCs) and multiple myeloma (MM) cells play crucial roles in MM development. We previously reported that BMSC-derived exosomes directly promote MM cell growth, whereas the other possible mechanisms for supporting MM progression by these exosomes are still not clear. Here, we investigated the effect of BMSC-derived exosomes on the MM BM cells with specific emphasis on myeloid-derived suppressor cells (MDSCs). BMSC-derived exosomes were able to be taken up by MM MDSCs and induced their expansion in vitro. Moreover, these exosomes directly induced the survival of MDSCs through activating STAT3 and STAT1 pathways and increasing the anti-apoptotic proteins Bcl-xL and Mcl-1. Inhibition of these pathways blocked the enhancement of MDSC survival. Furthermore, these exosomes increased the nitric oxide release from MM MDSCs and enhanced their suppressive activity on T cells. Taken together, our results demonstrate that BMSC-derived exosomes activate MDSCs in the BM through STAT3 and STAT1 pathways, leading to increased immunosuppression which favors MM progression.
Collapse
Affiliation(s)
- Jinheng Wang
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Nathan De Beule
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Ken Maes
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Elke De Bruyne
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Els Van Valckenborgh
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Karin Vanderkerken
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| |
Collapse
|
39
|
Takeda Y, Kato T, Ito H, Kurota Y, Yamagishi A, Sakurai T, Araki A, Nara H, Tsuchiya N, Asao H. The pattern of GPI-80 expression is a useful marker for unusual myeloid maturation in peripheral blood. Clin Exp Immunol 2016; 186:373-386. [PMID: 27569996 DOI: 10.1111/cei.12859] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2016] [Indexed: 02/06/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) have a wide spectrum of immunosuppressive activity; control of these cells is a new target for improving clinical outcomes in cancer patients. MDSCs originate from unusual differentiation of neutrophils or monocytes induced by inflammatory cytokines, including granulocyte-colony stimulating factor (G-CSF) and granulocyte-macrophage (GM)-CSF. However, MDSCs are difficult to detect in neutrophil or monocyte populations because they are not uniform cells, resembling both neutrophils and monocytes; thus, they exist in a heterogeneous population. In this study, we investigated GPI-80, a known regulator of Mac-1 (CD11b/CD18) and associated closely with neutrophil maturation, to clarify this unusual differentiation. First, we demonstrated that the mean fluorescence intensity (MFI) of GPI-80 and coefficient of variation (CV) of GPI-80 were increased by treatment with G-CSF and GM-CSF, respectively, using a human promyelocytic leukaemia (HL60) cell differentiation model. To confirm the value of GPI-80 as a marker of unusual differentiation, we measured GPI-80 expression and MDSC functions using peripheral blood cells from metastatic renal cell carcinoma patients. The GPI-80 CV was augmented significantly in the CD16hi neutrophil cell population, and GPI-80 MFI was increased significantly in the CD33hi monocyte cell population. Furthermore, the GPI-80 CV in the CD16hi population was correlated inversely with the proliferative ability of T cells and the GPI-80 MFI of the CD33hi population was correlated with reactive oxygen species production. These results led us to propose that the pattern of GPI-80 expression in these populations is a simple and useful marker for unusual differentiation, which is related to MDSC functions.
Collapse
Affiliation(s)
- Y Takeda
- Department of Immunology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - T Kato
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - H Ito
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - Y Kurota
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - A Yamagishi
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - T Sakurai
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - A Araki
- Department of Immunology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - H Nara
- Department of Immunology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - N Tsuchiya
- Department of Urology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| | - H Asao
- Department of Immunology, Yamagata University, Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
40
|
Wang J, De Veirman K, Faict S, Frassanito MA, Ribatti D, Vacca A, Menu E. Multiple myeloma exosomes establish a favourable bone marrow microenvironment with enhanced angiogenesis and immunosuppression. J Pathol 2016; 239:162-73. [PMID: 26956697 DOI: 10.1002/path.4712] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 02/03/2016] [Accepted: 02/29/2016] [Indexed: 12/21/2022]
Abstract
Multiple myeloma (MM) pathogenesis and progression largely rely on the cells and extracellular factors in the bone marrow (BM) microenvironment. Compelling studies have identified tumour exosomes as key regulators in the maintenance and education of the BM microenvironment by targeting stromal cells, immune cells, and vascular cells. However, the role of MM exosomes in the modification of the BM microenvironment and MM progression remains unclear. Here, we explored the functions of MM exosomes in angiogenesis and immunosuppression in vitro and in vivo. Murine MM exosomes carrying multiple angiogenesis-related proteins enhanced angiogenesis and directly promoted endothelial cell growth. Several pathways such as signal transducer and activator of transcription 3 (STAT3), c-Jun N-terminal kinase, and p53 were modulated by the exosomes in endothelial and BM stromal cells. These exosomes promoted the growth of myeloid-derived suppressor cells (MDSCs) in naive mice through activation of the STAT3 pathway and changed their subsets to similar phenotypes to those seen in MM-bearing mice. Moreover, MM exosomes up-regulated inducible nitric oxide synthase and enhanced the immunosuppressive capacity of BM MDSCs in vivo. Our data show that MM exosomes modulate the BM microenvironment through enhancement of angiogenesis and immunosuppression, which will further facilitate MM progression. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jinheng Wang
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Kim De Veirman
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Sylvia Faict
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| | - Maria Antonia Frassanito
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy.,National Cancer Institute 'Giovanni Paolo II', Bari, Italy
| | - Angelo Vacca
- Department of Biomedical Sciences and Human Oncology, University of Bari Medical School, Bari, Italy
| | - Eline Menu
- Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussels (VUB), Brussels, Belgium
| |
Collapse
|
41
|
Yu GT, Bu LL, Zhao YY, Mao L, Deng WW, Wu TF, Zhang WF, Sun ZJ. CTLA4 blockade reduces immature myeloid cells in head and neck squamous cell carcinoma. Oncoimmunology 2016; 5:e1151594. [PMID: 27471622 DOI: 10.1080/2162402x.2016.1151594] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 01/16/2023] Open
Abstract
Immature myeloid cells such as myeloid-derived suppressor cells (MDSCs) and M2 macrophages play a vital role in the tumor immune escape and tumor progression. Cytotoxic T lymphocyte-associated antigen 4 (CTLA4), as a negative immune checkpoint, is highly expressed in numerous solid tumors. However, precise functions of CTLA4 in head and neck squamous cell carcinoma (HNSCC) have not yet been elucidated. In this study, we demonstrated that the ratio of CD8(+)/CTLA4 can be used as a potential index with a clinical prognostic value for HNSCC. Using immunocompetent transgenic mouse model with spontaneous HNSCC, we directly observed that targeting CTLA4 decreases MDSCs and M2 macrophages and promotes T cell activation in both tumor microenvironment and macro-environment. In all, our study provides direct evidence in vivo and proposes a rationale for CTLA4 inhibition as a future therapeutic strategy in patients with HNSCC.
Collapse
Affiliation(s)
- Guang-Tao Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education , Wuhan, China
| | - Lin-Lin Bu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education , Wuhan, China
| | - Yu-Yue Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education , Wuhan, China
| | - Liang Mao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education , Wuhan, China
| | - Wei-Wei Deng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education , Wuhan, China
| | - Tian-Fu Wu
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University , Wuhan, China
| | - Wen-Feng Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University , Wuhan, China
| | - Zhi-Jun Sun
- The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine, Ministry of Education, Wuhan, China; Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
42
|
Li J, Srivastava RM, Ettyreddy A, Ferris RL. Cetuximab ameliorates suppressive phenotypes of myeloid antigen presenting cells in head and neck cancer patients. J Immunother Cancer 2015; 3:54. [PMID: 26579227 PMCID: PMC4647471 DOI: 10.1186/s40425-015-0097-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 10/23/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Myeloid-derived suppressor cells (MDSC) and M2 monocytes/macrophages are two types of suppressive myeloid antigen presenting cells that have been shown to promote tumor progression and correlate with poor prognosis in cancer patients. Tumor antigen specific monoclonal antibodies (mAb) have emerged as important agents for cancer therapy. In addition to the direct inhibition of tumor growth, the Fc portions of the therapeutic mAbs, such as the IgG1 portion of the anti-epidermal growth factor receptor (EGFR) mAb cetuximab, might interact with the Fc-gamma receptors (FcγR) on myeloid cells and modulate their suppressive activity. METHODS Patients with locally advanced head and neck squamous cell carcinoma (HNSCC) on the UPCI 08-013 NCT01218048 trial were treated with single-agent cetuximab before surgery. Blood were collected pre- and post-cetuximab treatment to analyze frequency of monocytic MDSC (CD11b(+)CD14(+)HLA-DR(lo/-)), granulocytic MDSC (LIN(-)CD11b(+)CD15(+)) and CD11b(+)CD14(+)HLA-DR(hi) monocytes by flow cytometry. Besides, CD11b(+)CD14(+)HLA-DR(hi) monocytes were sorted for qPCR analysis of IL-10 and IL-12B transcripts. MDSC were generated in vitro with or without coated hIgG1 and tested for suppressive activity in mixed leukocyte reaction (MLR). Naïve monocytes from HNSCC patients co-cultured with tumor cell lines in the presence of cetuximab or hIgG1 were analyzed for M1/2 surface markers and cytokines. RESULTS We observed significantly increased monocytic MDSC in non-responders and decreased granulocytic MDSC in responders after cetuximab treatment. In addition, circulating CD11b(+)CD14(+)HLA-DR(hi) monocytes of cetuximab responders displayed attenuated M2 polarization, with decreased CD163(+) expression and IL-10 transcripts after cetuximab treatment. This beneficial effect appeared to be FcγR dependent, since CD16 ligation reproduced the reversal of suppressive activity of MDSC in vitro. CD14(+) naïve monocytes from the co-cultures of tumor cells, cetuximab and HNSCC patient PBMC or purified monocytes were skewed to an M1-like phenotype, with increased expression of HLA-DR, CD86 and production of IL-12 p70. Likewise, reduced M2 features (expression of CD163 and production of IL-10) were found after crosslinking CD16 on the surface of monocytes to cetuximab-coated tumor cells. CONCLUSION Our studies demonstrate a novel function of cetuximab in ameliorating suppressive phenotypes of FcγR bearing myeloid cells in cancer patients, which is associated with better clinical outcome of cetuximab-treated patients. CLINICAL TRIAL REGISTRY #NCT01218048. Registered 7 October 2010.
Collapse
Affiliation(s)
- Jing Li
- />Department of Pharmacology and Pharmaceutical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | | | | | - Robert L. Ferris
- />Department of Otolaryngology, University of Pittsburgh, Pittsburgh, PA USA
- />Department of Immunology, University of Pittsburgh, Pittsburgh, PA USA
- />Cancer Immunology Program, University of Pittsburgh Cancer Institute, Pittsburgh, PA USA
- />Hillman Cancer Center Research Pavilion, 5117 Centre Avenue, Room 2.26b, Pittsburgh, PA 15213-1863 USA
| |
Collapse
|
43
|
Poisson LM, Suhail H, Singh J, Datta I, Denic A, Labuzek K, Hoda MN, Shankar A, Kumar A, Cerghet M, Elias S, Mohney RP, Rodriguez M, Rattan R, Mangalam AK, Giri S. Untargeted Plasma Metabolomics Identifies Endogenous Metabolite with Drug-like Properties in Chronic Animal Model of Multiple Sclerosis. J Biol Chem 2015; 290:30697-712. [PMID: 26546682 DOI: 10.1074/jbc.m115.679068] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Indexed: 12/20/2022] Open
Abstract
We performed untargeted metabolomics in plasma of B6 mice with experimental autoimmune encephalitis (EAE) at the chronic phase of the disease in search of an altered metabolic pathway(s). Of 324 metabolites measured, 100 metabolites that mapped to various pathways (mainly lipids) linked to mitochondrial function, inflammation, and membrane stability were observed to be significantly altered between EAE and control (p < 0.05, false discovery rate <0.10). Bioinformatics analysis revealed six metabolic pathways being impacted and altered in EAE, including α-linolenic acid and linoleic acid metabolism (PUFA). The metabolites of PUFAs, including ω-3 and ω-6 fatty acids, are commonly decreased in mouse models of multiple sclerosis (MS) and in patients with MS. Daily oral administration of resolvin D1, a downstream metabolite of ω-3, decreased disease progression by suppressing autoreactive T cells and inducing an M2 phenotype of monocytes/macrophages and resident brain microglial cells. This study provides a proof of principle for the application of metabolomics to identify an endogenous metabolite(s) possessing drug-like properties, which is assessed for therapy in preclinical mouse models of MS.
Collapse
Affiliation(s)
- Laila M Poisson
- From the Center for Bioinformatics and Departments of Public Health Sciences and
| | | | | | - Indrani Datta
- From the Center for Bioinformatics and Departments of Public Health Sciences and
| | | | - Krzysztof Labuzek
- the Department of Pharmacology, Medical University of Silesia, Medyków 18, PL 40-752 Katowice, Poland
| | - Md Nasrul Hoda
- the Department of Neurology, Georgia Health Sciences University, Augusta, Georgia 30912, the Program in Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, Georgia 30912
| | | | - Ashok Kumar
- the Department of Anatomy and Cell Biology, School of Medicine, Wayne State University, Detroit, Michigan 48202
| | | | | | | | - Moses Rodriguez
- the Departments of Neurology and Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota 55906
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Health System, Detroit, Michigan 48202
| | - Ashutosh K Mangalam
- the Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242
| | | |
Collapse
|
44
|
Guan Q, Blankstein AR, Anjos K, Synova O, Tulloch M, Giftakis A, Yang B, Lambert P, Peng Z, Cuvelier GD, Wall DA. Functional Myeloid-Derived Suppressor Cell Subsets Recover Rapidly after Allogeneic Hematopoietic Stem/Progenitor Cell Transplantation. Biol Blood Marrow Transplant 2015; 21:1205-14. [DOI: 10.1016/j.bbmt.2015.04.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 04/09/2015] [Indexed: 12/15/2022]
|
45
|
Xiao P, Wan X, Cui B, Liu Y, Qiu C, Rong J, Zheng M, Song Y, Chen L, He J, Tan Q, Wang X, Shao X, Liu Y, Cao X, Wang Q. Interleukin 33 in tumor microenvironment is crucial for the accumulation and function of myeloid-derived suppressor cells. Oncoimmunology 2015; 5:e1063772. [PMID: 26942079 DOI: 10.1080/2162402x.2015.1063772] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 06/11/2015] [Accepted: 06/12/2015] [Indexed: 12/31/2022] Open
Abstract
Tumor-induced, myeloid-derived suppressor cells (MDSCs)-mediated immune dysfunction is an important mechanism that leads to tumor immune escape and the inefficacy of cancer immunotherapy. Importantly, tumor-infiltrating MDSCs have much stronger ability compared to MDSCs in the periphery. However, the mechanisms that tumor microenvironment induces the accumulation and function of MDSCs are poorly understood. Here, we report that Interleukin-33 (IL-33) - a cytokine which can be abundantly released in tumor tissues both in 4T1-bearing mice and breast cancer patients, is crucial for facilitating the expansion of MDSCs. IL-33 in tumor microenvironment reduces the apoptosis and sustains the survival of MDSCs through induction of autocrine secretion of GM-CSF, which forms a positive amplifying loop for MDSC accumulation. This is in conjunction with IL-33-driven induction of arginase-1 expression and activation of NF-κB and MAPK signaling in MDSCs which augments their immunosuppressive ability, and histone modifications were involved in IL-33 signaling in MDSCs. In ST2-/- mice, the defect of IL-33 signaling in MDSCs attenuates the immunosuppressive and pro-tumoral capacity of MDSCs. Our results identify IL-33 as a critical mediator that contributes to the abnormal expansion and enhanced immunosuppressive function of MDSCs within tumor microenvironment, which can be potentially targeted to reverse MDSC-mediated tumor immune evasion.
Collapse
Affiliation(s)
- Peng Xiao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China; Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaopeng Wan
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Bijun Cui
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Yang Liu
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Chenyang Qiu
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Jiabing Rong
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Mingzhu Zheng
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Yinjing Song
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Luoquan Chen
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Jia He
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | - Qinchun Tan
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| | | | | | - Yuhua Liu
- The affiliated hospital of Hangzhou Normal University , Hangzhou, China
| | - Xuetao Cao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China; National Key Laboratory of Medical Molecular Biology and Department of Immunology, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine , Hangzhou, China
| |
Collapse
|
46
|
The Role and Potential Therapeutic Application of Myeloid-Derived Suppressor Cells in Allo- and Autoimmunity. Mediators Inflamm 2015; 2015:421927. [PMID: 26078493 PMCID: PMC4452474 DOI: 10.1155/2015/421927] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 04/21/2015] [Accepted: 04/28/2015] [Indexed: 12/16/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that consists of myeloid progenitor cells and immature myeloid cells. They have been identified as a cell population that may affect the activation of CD4(+) and CD8(+) T-cells to regulate the immune response negatively, which makes them attractive targets for the treatment of transplantation and autoimmune diseases. Several studies have suggested the potential suppressive effect of MDSCs on allo- and autoimmune responses. Conversely, MDSCs have also been found at various stages of differentiation, accumulating during pathological situations, not only during tumor development but also in a variety of inflammatory immune responses, bone marrow transplantation, and some autoimmune diseases. These findings appear to be contradictory. In this review, we summarize the roles of MDSCs in different transplantation and autoimmune diseases models as well as the potential to target these cells for therapeutic benefit.
Collapse
|
47
|
Pan W, Sun Q, Wang Y, Wang J, Cao S, Ren X. Highlights on mechanisms of drugs targeting MDSCs: providing a novel perspective on cancer treatment. Tumour Biol 2015; 36:3159-69. [DOI: 10.1007/s13277-015-3363-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/19/2015] [Indexed: 12/22/2022] Open
|
48
|
Ochando JC, Conde P. Editorial: Dexamethasone and MDSC in transplantation: yes to NO. J Leukoc Biol 2015; 96:669-71. [PMID: 25360039 DOI: 10.1189/jlb.3ce0514-272r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- J C Ochando
- Department of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - P Conde
- Department of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
49
|
Béland S, Désy O, Vallin P, Basoni C, De Serres SA. Innate immunity in solid organ transplantation: an update and therapeutic opportunities. Expert Rev Clin Immunol 2015; 11:377-89. [PMID: 25644774 DOI: 10.1586/1744666x.2015.1008453] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Innate immunity is increasingly recognized as a major player in transplantation. In addition to its role in inflammation in the early post-transplant period, innate immunity shapes the differentiation of cells of adaptive immunity, with a capacity to promote either rejection or tolerance. Emerging data indicate that innate allorecognition, a characteristic previously limited to lymphocytes, is involved in allograft rejection. This review briefly summarizes the physiology of each component of the innate immune system in the context of transplantation and presents the current or promising therapeutic applications, such as cellular, anticomplement and anticytokine therapies.
Collapse
Affiliation(s)
- Stéphanie Béland
- Transplantation Unit, Renal Division, Department of Medicine, CHU de Québec Research Center, Faculty of Medicine, Laval University, 11 Côte du Palais, Québec, QC, Canada
| | | | | | | | | |
Collapse
|
50
|
Bhatt S, Shen GQ, Li Y, Qian S, Ragni MV, Lu L. Hepatic stellate cell-conditioned myeloid cells provide a novel therapy for prevention of factor VIII antibody formation in mice. Exp Hematol 2014; 43:277-85. [PMID: 25534204 DOI: 10.1016/j.exphem.2014.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 11/11/2014] [Accepted: 12/03/2014] [Indexed: 12/14/2022]
Abstract
A major complication of factor VIII (F.VIII) infusion therapies for the treatment of hemophilia A is the formation of antibodies (inhibitors) against F.VIII, a T-cell-dependent, B-cell-mediated process. To date, attempts to inhibit formation of the inhibitors have been limited in success. We have shown that hepatic stellate cells (HSCs) promote the development of myeloid-derived suppressor cells (MDSCs). The HSC-induced MDSCs are potent regulators of T-cell and B-cell responses. Here we show that MDSCs can be propagated from hemophilia A mouse bone marrow cells in coculture with HSCs. These cells exhibit a suppressive phenotype and display a marked ability to inhibit T-cell proliferation induced by dendritic cells in response to F.VIII. MDSCs can also inhibit proliferation and activation of B cells stimulated by immunoglobulin M and interleukin 4. Administration of HSC-induced MDSCs induces CD4(+) T cell and B220(+) B-cell hyporesponsiveness to F.VIII and reduces inhibitor formation in hemophilia A mice. These results suggest that MDSCs could serve as a form of immunotherapy for preventing inhibitor formation via induction of immune tolerance.
Collapse
Affiliation(s)
- Sumantha Bhatt
- Department of Molecular Medicine, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States; Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Gong-Qing Shen
- Department of General Surgery, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Yan Li
- Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Shiguang Qian
- Department of Molecular Medicine, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States; Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States; Department of General Surgery, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Margaret V Ragni
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh and Hemophilia Center of Western Pennsylvania, Pittsburgh, PA, United States.
| | - Lina Lu
- Department of Molecular Medicine, Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, United States; Department of Immunology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States; Department of General Surgery, Digestive Disease Institute, Cleveland Clinic, Cleveland, OH, United States.
| |
Collapse
|