1
|
Wang R, Liu C, Liu X, Liu L, Xiao Y, Huo Y. Association between cellular immune and preeclampsia and preterm birth: A Mendelian randomization study. J Reprod Immunol 2024; 166:104391. [PMID: 39532026 DOI: 10.1016/j.jri.2024.104391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 09/03/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Emerging evidences have highlighted immune-inflammatory imbalances as a critical driver of the pathogenesis for preeclampsia (PE) and preterm birth (PB), but the impact of specific immune factors on the diseases is largely unknown. Our aim was to determine whether these immune cells are causally associated with the onset of PE or PB. Drawing on publicly available genetic data, we applied Mendelian randomization analysis to probe the causal link of 731 immune traits (7 panels: TBNK panel, Regulatory T cells panel, Maturation stages of T-cell panel, Dendritic cell panel, B-cell panel, Monocyte panel and Myeloid cell pane) with the risk of PE and PB. The inverse variance weighting method (IVW) acted as the primary analysis to estimate the validity of causality, and sensitivity analyses were conducted to assessment of heterogeneity and pleiotropy. After adjusting for P-values for FDR method, CD27 on CD24+ CD27+ B cell, CD80 on plasmacytoid Dendritic Cell, CD33+ HLA DR+ CD14dim Absolute Count, CD33+ HLA DR+ CD14- Absolute Count, CD33+ HLA DR+ Absolute Count were remarkably causally involved in increased risk of PE, while HLA DR on Dendritic Cell exerted a protective causation against PE (PFDR < 0.05). Moreover, we determined that CD45 on CD33dim HLA DR- in myeloid cells decreased PB risk, whereas CD11b on Granulocytic Myeloid-Derived Suppressor Cells had the opposite effect on PB (FFDR < 0.2). This study provided genetic evidence for causal relationships between immune cell traits and PE and PB, offering potential candidate immunophenotypes for future studies on the etiology of pregnancy complications.
Collapse
Affiliation(s)
- Runfang Wang
- Department of Obstetrics and Gynecology, Hebei General Hospital, Hebei 000050, China
| | - Cuilian Liu
- Department of Obstetrics and Gynecology, Hebei General Hospital, Hebei 000050, China
| | - Xiaodan Liu
- Department of Obstetrics and Gynecology, Hebei General Hospital, Hebei 000050, China
| | - Li Liu
- Department of Obstetrics and Gynecology, Hebei General Hospital, Hebei 000050, China
| | - Yuange Xiao
- Department of Obstetrics and Gynecology, Hebei General Hospital, Hebei 000050, China
| | - Yan Huo
- Department of Obstetrics and Gynecology, Hebei General Hospital, Hebei 000050, China.
| |
Collapse
|
2
|
Nieves C, Victoria da Costa Ghignatti P, Aji N, Bertagnolli M. Immune Cells and Infectious Diseases in Preeclampsia Susceptibility. Can J Cardiol 2024; 40:2340-2355. [PMID: 39304126 DOI: 10.1016/j.cjca.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/26/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024] Open
Abstract
Preeclampsia is a severe pregnancy disorder, affecting approximately 10% of pregnancies worldwide, characterised by hypertension and proteinuria after the 20th week of gestation. The condition poses significant risks to both maternal and fetal health, including cardiovascular complications and impaired fetal development. Recent trends indicate a rising incidence of preeclampsia, correlating with factors such as advanced maternal age and cardiovascular comorbidities. Emerging evidence also highlights a notable increase in the association between autoimmune and infectious diseases with preeclampsia. Autoimmune conditions, such as type 1 diabetes and systemic lupus erythematosus, and infections triggered by global health challenges, including leptospirosis, Zika, toxoplasmosis, and Chagas disease, are now recognised as significant contributors to preeclampsia susceptibility by affecting placental formation and function. This review focuses on the immunologic mechanisms underpinning preeclampsia, exploring how immune system dysregulation and infectious triggers exacerbate the condition. It also discusses the pathologic mechanisms, including galectins, that preeclampsia shares with autoimmune and infectious diseases, and their significant risk for adverse pregnancy outcomes. We emphasise the necessity for accurate diagnosis and vigilant monitoring of immune and infectious diseases during pregnancy to optimise management and reduce risks. By raising awareness about these evolving risks and their impact on pregnancy, we aim to enhance diagnostic practices and preventive strategies, ultimately improving outcomes for pregnant women, especially in regions affected by environmental changes and endemic diseases.
Collapse
Affiliation(s)
- Cecilia Nieves
- Cardiovascular Health Across the Lifespan Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; School of Physical and Occupational Therapy, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada.
| | - Paola Victoria da Costa Ghignatti
- Cardiovascular Health Across the Lifespan Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; School of Physical and Occupational Therapy, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada
| | - Narjiss Aji
- Cardiovascular Health Across the Lifespan Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Mariane Bertagnolli
- Cardiovascular Health Across the Lifespan Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada; School of Physical and Occupational Therapy, Faculty of Medicine and Health Sciences, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
3
|
Wang Y, Zhao X, Li Z, Wang W, Jiang Y, Zhang H, Liu X, Ren Y, Xu X, Hu X. Decidual natural killer cells dysfunction is caused by IDO downregulation in dMDSCs with Toxoplasma gondii infection. Commun Biol 2024; 7:669. [PMID: 38822095 PMCID: PMC11143278 DOI: 10.1038/s42003-024-06365-5] [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: 09/26/2023] [Accepted: 05/21/2024] [Indexed: 06/02/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) play a crucial role in maintaining maternal-fetal tolerance by expressing some immune-suppressive molecules, such as indoleamine 2,3-dioxygenase (IDO). Toxoplasma gondii (T. gondii) infection can break the immune microenvironment of maternal-fetal interface, resulting in adverse pregnancy outcomes. However, whether T. gondii affects IDO expression in dMDSCs and the molecular mechanism of its effect are still unclear. Here we show, the mRNA level of IDO is increased but the protein level decreased in infected dMDSCs. Mechanistically, the upregulation of transcriptional levels of IDO in dMDSCs is regulated through STAT3/p52-RelB pathway and the decrease of IDO expression is due to its degradation caused by increased SOCS3 after T. gondii infection. In vivo, the adverse pregnancy outcomes of IDO-/- infected mice are more severe than those of wide-type infected mice and obviously improved after exogenous kynurenine treatment. Also, the reduction of IDO in dMDSCs induced by T. gondii infection results in the downregulation of TGF-β and IL-10 expression in dNK cells regulated through Kyn/AhR/SP1 signal pathway, eventually leading to the dysfunction of dNK cells and contributing the occurrence of adverse pregnancy outcomes. This study reveals a novel molecular mechanism in adverse pregnancy outcome induced by T. gondii infection.
Collapse
Affiliation(s)
- Yu Wang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Xiaoyue Zhao
- Department of Clinical Psychology, Yantai Affiliated hospital of Binzhou Medial University, Yantai, 264100, Shandong, PR China
| | - Zhidan Li
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Wenxiao Wang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Yuzhu Jiang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Haixia Zhang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Xianbing Liu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Yushan Ren
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Xiaoyan Xu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, PR China.
| |
Collapse
|
4
|
Yao Y, Ye Y, Chen J, Zhang M, Cai X, Zheng C. Maternal-fetal immunity and recurrent spontaneous abortion. Am J Reprod Immunol 2024; 91:e13859. [PMID: 38722063 DOI: 10.1111/aji.13859] [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: 12/21/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 06/26/2024] Open
Abstract
Recurrent Spontaneous Abortion (RSA) is a common pregnancy complication, that has multifactorial causes, and currently, 40%-50% of cases remain unexplained, referred to as Unexplained RSA (URSA). Due to the elusive etiology and mechanisms, clinical management is exceedingly challenging. In recent years, with the progress in reproductive immunology, a growing body of evidence suggests a relationship between URSA and maternal-fetal immunology, offering hope for the development of tailored treatment strategies. This article provides an immunological perspective on the pathogenesis, diagnosis, and treatment of RSA. On one hand, it comprehensively reviews the immunological mechanisms underlying RSA, including abnormalities in maternal-fetal interface immune tolerance, maternal-fetal interface immune cell function, gut microbiota-mediated immune dysregulation, and vaginal microbiota-mediated immune anomalies. On the other hand, it presents the diagnosis and existing treatment modalities for RSA. This article offers a clear knowledge framework for understanding RSA from an immunological standpoint. In conclusion, while the "layers of the veil" regarding immunological factors in RSA are gradually being unveiled, our current research may only scratch the surface. In terms of immunological etiology, effective diagnostic tools for RSA are currently lacking, and the efficacy and safety of immunotherapies, primarily based on lymphocyte immunotherapy and intravenous immunoglobulin, remain contentious.
Collapse
Affiliation(s)
- Yao Yao
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Yiqing Ye
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Jia Chen
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Meng Zhang
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Xiaoyu Cai
- Department of Pharmacy, Hangzhou First People's Hospital, Hangzhou, China
| | - Caihong Zheng
- Department of Pharmacy, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| |
Collapse
|
5
|
Li Y, Guo J, Zhang H, Li Z, Ren Y, Jiang Y, Liu X, Hu X. LILRB4 regulates the function of decidual MDSCs via the SHP-2/STAT6 pathway during Toxoplasma gondii infection. Parasit Vectors 2023; 16:237. [PMID: 37461040 PMCID: PMC10353217 DOI: 10.1186/s13071-023-05856-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Toxoplasma gondii infection can cause adverse pregnancy outcomes, such as recurrent abortion, fetal growth restriction and infants with malformations, among others. Decidual myeloid-derived suppressor cells (dMDSCs) are a novel immunosuppressive cell type at the fetal-maternal interface which play an important role in sustaining normal pregnancy that is related to their high expression of the inhibitory molecule leukocyte immunoglobulin-like receptor B4 (LILRB4). It has been reported that the expression of LILRB4 is downregulated on decidual macrophages after T. gondii infection, but it remains unknown whether T. gondii infection can induce dMDSC dysfunction resulting from the change in LILRB4 expression. METHODS LILRB4-deficient (LILRB4-/-) pregnant mice infected with T. gondii with associated adverse pregnancy outcomes, and anti-LILRB4 neutralized antibodies-treated infected human dMDSCs were used in vivo and in vitro experiments, respectively. The aim was to investigate the effect of LILRB4 expression on dMDSC dysfunction induced by T. gondii infection. RESULTS Toxoplasma gondii infection was observed to reduce STAT3 phosphorylation, resulting in decreased LILRB4 expression on dMDSCs. The levels of the main functional molecules (arginase-1 [Arg-1], interleukin-10 [IL-10]) and main signaling molecules (phosphorylated Src-homology 2 domain-containing protein tyrosine phosphatase [p-SHP2], phosphorylated signal transducer and activator of transcription 6 [p-STAT6]) in dMDSCs were all significantly reduced in human and mouse dMDSCs due to the decrease of LILRB4 expression induced by T. gondii infection. SHP-2 was found to directly bind to STAT6 and STAT6 to bind to the promoter of the Arg-1 and IL-10 genes during T. gondii infection. CONCLUSIONS The downregulation of LILRB4 expression on dMDSCs induced by T. gondii infection could regulate the expression of Arg-1 and IL-10 via the SHP-2/STAT6 pathway, resulting in the dysfunction of dMDSCs, which might contribute to adverse outcomes during pregnancy by T. gondii infection.
Collapse
Affiliation(s)
- Yuantao Li
- Department of Gynecology and Obstetrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Jingjing Guo
- Department of Gynecology and Obstetrics, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, People's Republic of China
| | - Haixia Zhang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Zhidan Li
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Yushan Ren
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Yuzhu Jiang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Xianbing Liu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| |
Collapse
|
6
|
Hua S, Wang S, Cai J, Wu L, Cao Y. Myeloid-derived suppressor cells: Are they involved in gestational diabetes mellitus? Am J Reprod Immunol 2023:e13711. [PMID: 37157925 DOI: 10.1111/aji.13711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/13/2023] [Accepted: 04/21/2023] [Indexed: 05/10/2023] Open
Abstract
Gestational diabetes mellitus (GDM) is currently the most common metabolic complication during pregnancy, with an increasing prevalence worldwide. Maternal immune dysregulation might be partly responsible for the pathophysiology of GDM. Myeloid derived suppressor cells (MDSCs) are a heterogeneous population of cells, emerging as a new immune regulator with potent immunosuppressive capacity. Although the fate and function of these cells were primarily described in pathological conditions such as cancer and infection, accumulating evidences have spotlighted their beneficial roles in homeostasis and physiological conditions. Recently, several studies have explored the roles of MDSCs in the diabetic microenvironment. However, the fate and function of these cells in GDM are still unknown. The current review summarized the existing knowledges about MDSCs and their potential roles in diabetes during pregnancy in an attempt to highlight our current understanding of GDM-related immune dysregulation and identify areas where further study is required.
Collapse
Affiliation(s)
- Siyu Hua
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| | - Shanshan Wang
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jinyang Cai
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lamei Wu
- Department of Perinatal Healthcare, Huai'an District Maternity and Child Health Hospital, Huai'an, Jiangsu, China
| | - Yan Cao
- Nanjing Maternity and Child Health Care Institute, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Shah NK, Xu P, Shan Y, Chen C, Xie M, Li Y, Meng Y, Shu C, Dong S, He J. MDSCs in pregnancy and pregnancy-related complications: an update†. Biol Reprod 2023; 108:382-392. [PMID: 36504233 DOI: 10.1093/biolre/ioac213] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/15/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
Maternal-fetal immune tolerance is a process that involves complex interactions of the immune system, and myeloid-derived suppressor cells have emerged as one of the novel immunomodulator in the maintenance of maternal-fetal immune tolerance. Myeloid-derived suppressor cells are myeloid progenitor cells with immunosuppressive activities on both innate and adaptive cells through various mechanisms. Emerging evidence demonstrates the accumulation of myeloid-derived suppressor cells during healthy pregnancy to establish maternal-fetal immune tolerance, placentation, and fetal-growth process. By contrast, the absence or decreased myeloid-derived suppressor cells in pregnancy complications like preeclampsia, preterm birth, stillbirth, and recurrent spontaneous abortion have been reported. Here, we have summarized the origin, mechanisms, and functions of myeloid-derived suppressor cells during pregnancy along with the recent advancements in this dynamic field. We also shed light on the immunomodulatory activity of myeloid-derived suppressor cells, which can be a foundation for potential therapeutic manipulation in immunological pregnancy complications.
Collapse
Affiliation(s)
- Neelam Kumari Shah
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Peng Xu
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yanhong Shan
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Chen Chen
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Min Xie
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yan Li
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Yizi Meng
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Chang Shu
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Shuai Dong
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jin He
- Department of Obstetrics, Obstetrics and Gynaecology Center, The First Hospital of Jilin University, Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration & Transplantation of Ministry of Education, National-Local Joint Engineering Laboratory of Animal Models for Human Diseases, The First Hospital of Jilin University, Jilin University, Changchun, China
| |
Collapse
|
8
|
Polcz VE, Rincon JC, Hawkins RB, Barrios EL, Efron PA, Moldawer LL, Larson SD. TRAINED IMMUNITY: A POTENTIAL APPROACH FOR IMPROVING HOST IMMUNITY IN NEONATAL SEPSIS. Shock 2023; 59:125-134. [PMID: 36383390 PMCID: PMC9957873 DOI: 10.1097/shk.0000000000002054] [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] [Indexed: 11/17/2022]
Abstract
ABSTRACT Sepsis, a dysregulated host immune response to infection, is one of the leading causes of neonatal mortality worldwide. Improved understanding of the perinatal immune system is critical to improve therapies to both term and preterm neonates at increased risk of sepsis. Our narrative outlines the known and unknown aspects of the human immune system through both the immune tolerant in utero period and the rapidly changing antigen-rich period after birth. We will highlight the key differences in innate and adaptive immunity noted through these developmental stages and how the unique immune phenotype in early life contributes to the elevated risk of overwhelming infection and dysregulated immune responses to infection upon exposure to external antigens shortly after birth. Given an initial dependence on neonatal innate immune host responses, we will discuss the concept of innate immune memory, or "trained immunity," and describe several potential immune modulators, which show promise in altering the dysregulated immune response in newborns and improving resilience to sepsis.
Collapse
Affiliation(s)
- Valerie E Polcz
- Department of Surgery and Sepsis and Critical Illness Research Center, University of Florida College of Medicine, Gainesville, Florida
| | | | | | | | | | | | | |
Collapse
|
9
|
Pang B, Hu C, Li H, Nie X, Wang K, Zhou C, Yi H. Myeloidderived suppressor cells: Escorts at the maternal-fetal interface. Front Immunol 2023; 14:1080391. [PMID: 36817414 PMCID: PMC9932974 DOI: 10.3389/fimmu.2023.1080391] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/13/2023] [Indexed: 02/05/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a novel heterogenous group of immunosuppressive cells derived from myeloid progenitors. Their role is well known in tumors and autoimmune diseases. In recent years, the role and function of MDSCs during reproduction have attracted increasing attention. Improving the understanding of their strong association with recurrent implantation failure, pathological pregnancy, and neonatal health has become a focus area in research. In this review, we focus on the interaction between MDSCs and other cell types (immune and non-immune cells) from embryo implantation to postpartum. Furthermore, we discuss the molecular mechanisms that could facilitate the therapeutic targeting of MDSCs. Therefore, this review intends to encourage further research in the field of maternal-fetal interface immunity in order to identify probable pathways driving the accumulation of MDSCs and to effectively target their ability to promote embryo implantation, reduce pathological pregnancy, and increase neonatal health.
Collapse
Affiliation(s)
- Bo Pang
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Cardiology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Cong Hu
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huimin Li
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, China
| | - Xinyu Nie
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Reproductive Medicine Center, Prenatal Diagnosis Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Keqi Wang
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China.,Cardiology Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Chen Zhou
- General Department, First Hospital of Jilin University, Changchun, Jilin, China
| | - Huanfa Yi
- Central Laboratory, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
10
|
Shibata M, Nanno K, Yoshimori D, Nakajima T, Takada M, Yazawa T, Mimura K, Inoue N, Watanabe T, Tachibana K, Muto S, Momma T, Suzuki Y, Kono K, Endo S, Takenoshita S. Myeloid-derived suppressor cells: Cancer, autoimmune diseases, and more. Oncotarget 2022; 13:1273-1285. [PMID: 36395389 PMCID: PMC9671473 DOI: 10.18632/oncotarget.28303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Although cancer immunotherapy using immune checkpoint inhibitors (ICIs) has been recognized as one of the major treatment modalities for malignant diseases, the clinical outcome is not uniform in all cancer patients. Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells that possess various strong immunosuppressive activities involving multiple immunocompetent cells that are significantly accumulated in patients who did not respond well to cancer immunotherapies. We reviewed the perspective of MDSCs with emerging evidence in this review. Many studies on MDSCs were performed in malignant diseases. Substantial studies on the participation of MDSCs on non-malignant diseases such as chronic infection and autoimmune diseases, and physiological roles in obesity, aging, pregnancy and neonates have yet to be reported. With the growing understanding of the roles of MDSCs, variable therapeutic strategies and agents targeting MDSCs are being investigated, some of which have been used in clinical trials. More studies are required in order to develop more effective strategies against MDSCs.
Collapse
Affiliation(s)
- Masahiko Shibata
- 1Department of Comprehensive Cancer Treatment and Research at Aizu, Fukushima Medical University, Fukushima, Japan,2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan,Correspondence to:Masahiko Shibata, email:
| | - Kotaro Nanno
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,5Department of Surgery, Nippon Medical School, Tokyo, Japan
| | - Daigo Yoshimori
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,5Department of Surgery, Nippon Medical School, Tokyo, Japan
| | - Takahiro Nakajima
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan
| | - Makoto Takada
- 4Aizu Oncology Consortium, Fukushima, Japan,6Department of Surgery, Bange Kousei General Hospital, Fukushima, Japan
| | - Takashi Yazawa
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan
| | - Kousaku Mimura
- 3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan
| | - Norio Inoue
- 2Department of Surgery, Cancer Treatment Center, Aizu Chuo Hospital, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan
| | - Takafumi Watanabe
- 7Department of Obstetrics and Gynecology, Fukushima Medical University, Fukushima, Japan
| | | | - Satoshi Muto
- 9Department of Chest Surgery, Fukushima Medical University, Fukushima, Japan
| | - Tomoyuki Momma
- 3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan
| | - Yoshiyuki Suzuki
- 1Department of Comprehensive Cancer Treatment and Research at Aizu, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan,10Department of Radiation Oncology, Fukushima Medical University, Fukushima, Japan
| | - Koji Kono
- 1Department of Comprehensive Cancer Treatment and Research at Aizu, Fukushima Medical University, Fukushima, Japan,3Department of Gastrointestinal Tract Surgery, Fukushima Medical University, Fukushima, Japan,4Aizu Oncology Consortium, Fukushima, Japan
| | - Shungo Endo
- 11Department of Colorectoanal Surgery, Aizu Medical Center, Fukushima Medical University, Fukushima, Japan
| | | |
Collapse
|
11
|
Leonardo L, Kenangalem E, Poespoprodjo JR, Noviyanti R, Price RN, Anstey NM, Minigo G, Kho S. Increased circulating myeloid-derived suppressor cells in vivax malaria and severe falciparum malaria. Malar J 2022; 21:255. [PMID: 36068577 PMCID: PMC9446641 DOI: 10.1186/s12936-022-04268-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 08/16/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Circulating myeloid-derived-suppressor-cells (MDSC) with immunosuppressive function are increased in human experimental Plasmodium falciparum infection, but have not been studied in clinical malaria. METHODS Using flow-cytometry, circulating polymorphonuclear-MDSC were evaluated in cryopreserved samples from patients with uncomplicated Plasmodium vivax (n = 8) and uncomplicated (n = 4) and severe (n = 16) falciparum malaria from Papua, Indonesia. RESULTS The absolute number of circulating polymorphonuclear-MDSC were significantly elevated in severe falciparum malaria patients compared to controls (n = 10). Polymorphonuclear-MDSC levels in uncomplicated vivax malaria were also elevated to levels comparable to that seen in severe falciparum malaria. CONCLUSION Control of expansion of immunosuppressive MDSC may be important for development of effective immune responses in falciparum and vivax malaria.
Collapse
Affiliation(s)
- Leo Leonardo
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
| | - Enny Kenangalem
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
| | - Jeanne R Poespoprodjo
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
- Department of Pediatrics, University of Gadjah Mada, Yogyakarta, Indonesia
| | | | - Ric N Price
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- Centre for Tropical Medicine, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX37LJ, UK
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nicholas M Anstey
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
| | - Gabriela Minigo
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- College of Health and Human Sciences, Charles Darwin University, Darwin, NT, Australia
| | - Steven Kho
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia.
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia.
| |
Collapse
|
12
|
Wang Q, Zhang X, Li C, Xiong M, Bai W, Sun S, Chen C, Zhang X, Li M, Zhao A. Intracellular Lipid Accumulation Drives the Differentiation of Decidual Polymorphonuclear Myeloid-Derived Suppressor Cells via Arachidonic Acid Metabolism. Front Immunol 2022; 13:868669. [PMID: 35664000 PMCID: PMC9159278 DOI: 10.3389/fimmu.2022.868669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Decidual polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) are essential to immune tolerance during pregnancy. A reduction in the number of these cells is associated with unexplained recurrent pregnancy loss (URPL). In our previous study, we reported that PMN-MDSCs are a group of mature neutrophils that are activated by the decidua microenvironment. In the present study, we show that the decidua microenvironment induces substantial lipid accumulation in neutrophils during their differentiation to PMN-MDSCs. Lower levels of lipid accumulation are detected in PMN-MDSCs from URPL patients, and the amount of lipid in the PMN-MDSCs is positively correlated with the proportion of PMN-MDSCs. Next, we demonstrate that decidua-derived IL6 with the presence of arachidonic acid upregulates fatty acid-binding protein 5 (FABP5) via the phosphorylation of signal transducer and activator of transcription 3 (STAT3). Fy -60ABP5 then continuously stimulates intracellular lipid accumulation. Increased intracellular lipid accumulation mediates arachidonic acid metabolism, a pathway that is significantly activated by the induction of the decidua microenvironment, to stimulate the synthesis of prostaglandin E2 (PGE2) and finally induce the differentiation of PMN-MDSCs. To summarize, decidua-derived IL6 facilitates the differentiation of PMN-MDSCs from neutrophils via the pSTAT3/FABP5/PGE2 pathway. Defects in the process may result in impaired differentiation and dysfunction of PMN-MDSCs in URPL. These findings enhance our understanding of the physiological mechanisms of immune tolerance in pregnancy and provide therapeutic options for URPL.
Collapse
Affiliation(s)
- Qiaohong Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Xinyang Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Congcong Li
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Miao Xiong
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China.,Department of Obstetrics and Gynecology, Shanghai Jiao Tong University School of Medicine Affiliated Sixth People's Hospital, Shanghai, China
| | - Wenxin Bai
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Si Sun
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Chao Chen
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Xiaoxin Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Mingyang Li
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Aimin Zhao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| |
Collapse
|
13
|
Bizymi N, Georgopoulou A, Mastrogamvraki N, Matheakakis A, Gontika I, Fragiadaki I, Mavroudi I, Papadaki HA. Myeloid-Derived Suppressor Cells (MDSC) in the Umbilical Cord Blood: Biological Significance and Possible Therapeutic Applications. J Clin Med 2022; 11:jcm11030727. [PMID: 35160177 PMCID: PMC8836851 DOI: 10.3390/jcm11030727] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of myeloid cells that suppress immune responses in cancer, infection, and trauma. They mainly act by inhibiting T-cells, natural-killer cells, and dendritic cells, and also by inducing T-regulatory cells, and modulating macrophages. Although they are mostly associated with adverse prognosis of the underlying disease entity, they may display positive effects in specific situations, such as in allogeneic hematopoietic stem cell transplantation (HSCT), where they suppress graft-versus-host disease (GVHD). They also contribute to the feto-maternal tolerance, and in the fetus growth process, whereas several pregnancy complications have been associated with their defects. Human umbilical cord blood (UCB) is a source rich in MDSCs and their myeloid progenitor cells. Recently, a number of studies have investigated the generation, isolation, and expansion of UCB-MDSCs for potential clinical application associated with their immunosuppressive properties, such as GVHD, and autoimmune and inflammatory diseases. Given that a significant proportion of UCB units in cord blood banks are not suitable for clinical use in HSCT, they might be used as a significant source of MDSCs for research and clinical purposes. The current review summarizes the roles of MDSCs in the UCB, as well as their promising applications.
Collapse
Affiliation(s)
- Nikoleta Bizymi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (N.B.); (A.M.); (I.M.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Anthie Georgopoulou
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Natalia Mastrogamvraki
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Angelos Matheakakis
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (N.B.); (A.M.); (I.M.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
| | - Ioanna Gontika
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Irene Fragiadaki
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Irene Mavroudi
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (N.B.); (A.M.); (I.M.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
| | - Helen A. Papadaki
- Department of Haematology, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (N.B.); (A.M.); (I.M.)
- Haemopoiesis Research Laboratory, School of Medicine, University of Crete, 71003 Heraklion, Crete, Greece
- Public Cord Blood Bank of Crete, University Hospital of Heraklion, 71500 Heraklion, Crete, Greece; (A.G.); (N.M.); (I.G.); (I.F.)
- Correspondence: ; Tel.: +30-2810394637
| |
Collapse
|
14
|
Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol 2021; 21:485-498. [PMID: 33526920 PMCID: PMC7849958 DOI: 10.1038/s41577-020-00490-y] [Citation(s) in RCA: 948] [Impact Index Per Article: 237.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2020] [Indexed: 01/30/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are pathologically activated neutrophils and monocytes with potent immunosuppressive activity. They are implicated in the regulation of immune responses in many pathological conditions and are closely associated with poor clinical outcomes in cancer. Recent studies have indicated key distinctions between MDSCs and classical neutrophils and monocytes, and, in this Review, we discuss new data on the major genomic and metabolic characteristics of MDSCs. We explain how these characteristics shape MDSC function and could facilitate therapeutic targeting of these cells, particularly in cancer and in autoimmune diseases. Additionally, we briefly discuss emerging data on MDSC involvement in pregnancy, neonatal biology and COVID-19.
Collapse
Affiliation(s)
- Filippo Veglia
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | | |
Collapse
|
15
|
Li C, Chen C, Kang X, Zhang X, Sun S, Guo F, Wang Q, Kou X, Bai W, Zhao A. Decidua-derived granulocyte macrophage colony-stimulating factor induces polymorphonuclear myeloid-derived suppressor cells from circulating CD15+ neutrophils. Hum Reprod 2021; 35:2677-2691. [PMID: 33067638 DOI: 10.1093/humrep/deaa217] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/23/2020] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Do decidua-derived factors stimulate the conversion of circulating neutrophils to polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in early human pregnancy? SUMMARY ANSWER Circulating neutrophils can acquire PMN-MDSC-like phenotypes and function via phosphorylated signal transducer and activator of transcription 5/programmed death ligand 2 (pSTAT5/PD-L2) signalling after stimulation with decidua-derived granulocyte macrophage colony-stimulating factor (GM-CSF). WHAT IS KNOWN ALREADY PMN-MDSCs are an important immunoregulatory cell type in early pregnancy. Neutrophils are of high heterogeneity and plasticity and can polarize to immunosuppressive PMN-MDSCs upon stimulation. STUDY DESIGN, SIZE, DURATION For analysis of myeloid-derived suppressor cell (MDSC) subset proportions, 12 endometrium tissues and 12 peripheral blood samples were collected from non-pregnant women, and 40 decidua tissues and 16 peripheral blood samples were obtained from women with normal early pregnancy undergoing elective surgical pregnancy termination for nonmedical reasons with gestation age of 6-10 weeks. Twenty-nine decidua tissues were collected for isolation of CD15+ PMN-MDSCs. Twenty endometrium tissues and 30 decidua tissues were collected for cytokine analysis, immunohistochemistry or neutrophil stimulation. Peripheral blood samples were obtained from 36 healthy donors for isolation of CD3+ T cells and CD15+ neutrophils. PARTICIPANTS/MATERIALS, SETTING, METHODS The proportion of MDSC subsets in the decidua and peripheral blood of normal early pregnancy, endometrium and peripheral blood of non-pregnant women was analysed by flow cytometry. The phenotypes and function of decidual PMN-MDSCs and circulating neutrophils were compared by flow cytometry. Circulating neutrophils were stimulated with decidual explant supernatant (DES) and the phenotypes were measured by flow cytometry and immunofluorescence. The suppressive capacity of decidual PMN-MDSCs and DES-conditioned neutrophils was analysed by flow cytometry with or without anti-programmed cell death-1 (PD-1) antibody. Cytokines from DES and endometrial explant supernatant (EES) were detected by a Luminex assay. GM-CSF expression was determined by ELISA and immunohistochemistry. Neutrophils were stimulated with DES, EES, DES with anti-GM-CSF antibody or EES with GM-CSF. CD11b, lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), PD-L2 and pSTAT5 expression were measured by flow cytometry. MAIN RESULTS AND THE ROLE OF CHANCE The frequency of PMN-MDSCs was significantly increased in the decidua of early pregnancy compared with peripheral blood of non-pregnant women, the endometrium of non-pregnant women or peripheral blood during early pregnancy. Decidual PMN-MDSCs suppressed T-cell proliferation and cytokine production. Phenotypes of decidual PMN-MDSCs were similar to mature activated neutrophils. DES-induced CD11b, LOX-1, PD-L2 expression and STAT5 phosphorylation in neutrophils. The PD-L2 expression in neutrophils was dependent on STAT5 phosphorylation. Both decidual PMN-MDSCs and DES-conditioned neutrophils suppressed T-cell proliferation via PD-1 signalling. GM-CSF was up-regulated in the decidua and induced CD11b, LOX-1 and PD-L2 expression on neutrophils. DES significantly induced CD11b, LOX-1, PD-L2 expression and STAT5 phosphorylation. Anti-GM-CSF antibody remarkably blocked such stimulation in neutrophils. EES did not induce CD11b, LOX-1, PD-L2 expression or STAT5 phosphorylation, while GM-CSF treatment sufficiently stimulated CD11b, LOX-1, PD-L2 expression and STAT5 phosphorylation in neutrophils. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The study was based on in vitro experiments and we were not able to evaluate neutrophils differentiation to PMN-MDSCs in other sites before entering the maternal-foetal interface due to the limited availability of human samples. This needs to be explored using murine models. WIDER IMPLICATIONS OF THE FINDINGS This is the first study demonstrating that decidual PMN-MDSCs are a group of immunoregulatory cells with mature status, and that neutrophils can be induced to a PMN-MDSC-like phenotype with decidua-derived GM-CSF via pSTAT5/PD-L2 signalling. This study indicates that GM-CSF can facilitate immune tolerance of early pregnancy through regulating PMN-MDSCs and further provides a potential role of GM-CSF in prevention and treatment for pregnancy complications. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Natural Science Foundation of China (81671481) and National Natural Science Foundation of China (81871179). All authors have no competing interests to declare.
Collapse
Affiliation(s)
- Congcong Li
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, PR China
| | - Chao Chen
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, PR China
| | - Xiaomin Kang
- Department of Reproductive Medical Center, The First People's Hospital of Yunnan Province, Kunming, Yunnan, PR China
| | - Xiaoxin Zhang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Si Sun
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Feng Guo
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, PR China
| | - Qiaohong Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, PR China
| | - Xi Kou
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Wenxin Bai
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China
| | - Aimin Zhao
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, PR China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, PR China
| |
Collapse
|
16
|
Chen Z, Zhang X, Lv S, Xing Z, Shi M, Li X, Chen M, Zuo S, Tao Y, Xiao G, Liu J, He Y. Treatment With Endothelin-A Receptor Antagonist BQ123 Attenuates Acute Inflammation in Mice Through T-Cell-Dependent Polymorphonuclear Myeloid-Derived Suppressor Cell Activation. Front Immunol 2021; 12:641874. [PMID: 33828553 PMCID: PMC8019801 DOI: 10.3389/fimmu.2021.641874] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/08/2021] [Indexed: 11/30/2022] Open
Abstract
The endothelin-A receptor antagonist BQ123 is an effective treatment agent for hypertension and obese cardiomyopathy. However, the role of BQ123 in controlling acute inflammatory diseases and its underlying mechanisms are not well understood. Here, we showed that BQ123 activated polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) in mice and that the IL13/STAT6/Arg1 signaling pathway is involved in this process. Importantly, both treatment with BQ123 and the transfer of BQ123-induced PMN-MDSCs (BQ123-MDSCs) were effective in relieving inflammation, including dextran sulfate sodium (DSS)-induced colitis, papain-induced pneumonia, and concanavalin A (ConA)-induced hepatitis, in mice. The treatment effects were mediated by the attenuation of the inflammation associated with the accumulation of PMN-MDSCs in the colon, lung, and liver. However, concurrent injection of Gr1 agonistic antibody with BQ123 induced PMN-MDSC aggravated the observed acute inflammation. Interestingly, no remission of inflammation was observed in Rag2 knockout mice administered BQ123-MDSCs, but co-injection with CD3+ T cells significantly relieved acute inflammation. In summary, BQ123-induced PMN-MDSCs attenuated acute inflammation in a T cell-dependent manner, providing a novel potential strategy to prevent the occurrence of acute inflammation.
Collapse
Affiliation(s)
- Ziyang Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaogang Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shuaijun Lv
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhe Xing
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Mengyu Shi
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xinyao Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Meiqi Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Shaowen Zuo
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yingxu Tao
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Gang Xiao
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Jingping Liu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Yumei He
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University, Guangzhou, China
| |
Collapse
|
17
|
Liu J, Yang Q, Chen Z, Lv S, Tang J, Xing Z, Shi M, Lei A, Xiao G, He Y. TFF3 mediates the NF-κB/COX2 pathway to regulate PMN-MDSCs activation and protect against necrotizing enterocolitis. Eur J Immunol 2021; 51:1110-1125. [PMID: 33547649 DOI: 10.1002/eji.202048768] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/27/2020] [Accepted: 02/04/2021] [Indexed: 12/17/2022]
Abstract
Intestinal trefoil factor 3 (TFF3) plays an important role in repairing the intestinal mucosa. However, the detailed mechanism regarding immune regulation by TFF3 is not well defined. Here, we reported that treatment of mouse BM cells and human peripheral blood mononuclear cells from healthy volunteers with TFF3 activated polymorphnuclear myeloid-derived suppressor cells (PMN-MDSCs) in vitro. We also found that prostaglandin E2 is a major TFF3-mediated MDSC target, and that NF-κB/COX2 signaling was involved in this process. Moreover, TFF3 treatment or transfer of TFF3-derived PMN-MDSCs (TFF3-MDSCs) to experimental necrotizing enterocolitis (NEC) mice caused PMN-MDSC accumulation in the lamina propria (LP), which was associated with decreased intestinal inflammation, permeability, bacterial loading, and prolonged survival. Interestingly, no NEC severity remission was observed in Rag1 KO mice that were given TFF3-MDSCs, but coinjection with CD4+ T cells significantly relieved NEC inflammation. Overall, TFF3 mediates the NF-κB/COX2 pathway to regulate PMN-MDSC activation and attenuates NEC in a T-cell-dependent manner, which suggests a novel mechanism in preventing NEC occurrence.
Collapse
Affiliation(s)
- Jingping Liu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, P. R. China
| | - Qiong Yang
- Chronic Disease Laboratory, Institutes for Life Sciences and School of Medicine, South China University of Technology, Guangzhou, P. R. China
| | - Ziyang Chen
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Shuaijun Lv
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Jian Tang
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhe Xing
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Mengyu Shi
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China
| | - Aihua Lei
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, University of South China, Hengyang, P. R. China
| | - Gang Xiao
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, P. R. China
| | - Yumei He
- Department of Clinical Laboratory, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, P. R. China.,Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, P. R. China.,Guangdong Provincial Key Laboratory of Proteomics, Southern Medical University, Guangzhou, P. R. China
| |
Collapse
|
18
|
Jiang HH, Wang KX, Bi KH, Lu ZM, Zhang JQ, Cheng HR, Zhang MY, Su JJ, Cao YX. Sildenafil might impair maternal-fetal immunotolerance by suppressing myeloid-derived suppressor cells in mice. J Reprod Immunol 2020; 142:103175. [PMID: 32682164 DOI: 10.1016/j.jri.2020.103175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 06/24/2020] [Accepted: 07/01/2020] [Indexed: 12/11/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) as an important population of immune cells were found to restrain T cell function, polarize T-helper cells (Th) 1/Th2 toward Th2 response and induce regulatory T cells (Tregs), therefore enhancing the immunotolerance during pregnancy. Sildenafil has been applied for poor endometrial quality in implantation failure patients. Nevertheless, investigations have shown that sildenafil could reduce MDSCs-dependent immunosuppression. Whether sildenafil affects embryo implantation by suppressing MDSCs? To address this question, using the mice model, we investigated the amounts of immune cells in peripheral blood and endometrial cells from control group (CG), sildenafil low-dose group (LDG) and high-dose group (HDG). We found that both treatment groups displayed a marked deficiency in polymorphonuclear (PMN)-MDSCs and Th2 from mice blood and endometrium as compared to these from CG. The frequency of Tregs in endometrium from HDG was lower than those from CG. Th1/Th2 ratio in both periphery and uterus from study groups showed a significant increase as compared to those from CG. By relevance analysis, we found that the level of Tregs positively correlated with the level of PMN-MDSCs, whereas the Th1/Th2 ratio negatively correlated with the frequency of PMN-MDSCs in uterus. Moreover, there was a positive relationship between the amount of blood PMN-MDSCs and endometrial PMN-MDSCs. These results suggest that we should carefully weigh the pros and cons of using sildenafil when applied to patients with poor endometrial receptivity.
Collapse
Affiliation(s)
- H H Jiang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - K X Wang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - K H Bi
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Z M Lu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - J Q Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - H R Cheng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - M Y Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - J J Su
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China
| | - Y X Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No 81 Meishan Road, Hefei 230032, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei 230032, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No 81 Meishan Road, Hefei 230032, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, No 81 Meishan Road, Hefei 230032, Anhui, China.
| |
Collapse
|
19
|
Zheng ZM, Yang HL, Lai ZZ, Wang CJ, Yang SL, Li MQ, Shao J. Myeloid-derived suppressor cells in obstetrical and gynecological diseases. Am J Reprod Immunol 2020; 84:e13266. [PMID: 32418253 DOI: 10.1111/aji.13266] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 12/15/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of myeloid-origin cells which have immunosuppressive activities in several conditions, such as cancer and inflammation. Recent research has also associated MDSCs with numerous obstetrical and gynecological diseases. During pregnancy, MDSCs accumulate to ensure maternal-fetal immune tolerance, whereas they are decreased in patients who suffer from early miscarriage or pre-eclampsia. While the etiology of endometriosis is still unknown, abnormal accumulation of MDSCs in the peripheral blood and peritoneal fluid, alongside an increased level of reactive oxygen species (ROS), has been observed in these patients, which is central to the cellular immune regulations by MDSCs. Additionally, the regulation of MDSCs observed in tumours is also applicable to gynecologic neoplasms, including ovarian cancer and cervical cancer. More recently, emerging evidence has shown that there are high levels of MDSCs in premature ovarian failure (POF) and in vitro fertilization (IVF), but the underlying mechanisms are unknown. In this review, the generation and mechanisms of MDSCs are summarized. In particular, the modulation of these cells in immune-related obstetrical and gynecological diseases is discussed, including potential treatment options targeting MDSCs.
Collapse
Affiliation(s)
- Zi-Meng Zheng
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Hui-Li Yang
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Zhen-Zhen Lai
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Cheng-Jie Wang
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Shao-Liang Yang
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Ming-Qing Li
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| | - Jun Shao
- Insitute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Cao P, Sun Z, Feng C, Zhang J, Zhang F, Wang W, Zhao Y. Myeloid-derived suppressor cells in transplantation tolerance induction. Int Immunopharmacol 2020; 83:106421. [PMID: 32217462 DOI: 10.1016/j.intimp.2020.106421] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 02/29/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous cells derived from bone marrow. These cells are developed from immature myeloid cells and have strong negative immunomodulatory effects. In the context of pathology (such as tumor, autoimmune disease, trauma, and burns), MDSCs accumulate around tumor and inflammatory tissues, where their main role is to inhibit the function of effector T cells and promote the recruitment of regulatory T cells. MDSCs can be used in organ transplantation to regulate the immune responses that participate in rejection of the transplanted organ. This effect is achieved by increasing the production of MDSCs in vivo or transfusion of MDSCs induced in vitro to establish immune tolerance and prolong the survival of the graft. In this review, we discuss the efficacy of MDSCs in a variety of transplantation studies as well as the induction of immune tolerance to prevent transplant rejection through the use of common clinical immunosuppressants combined with MDSCs.
Collapse
Affiliation(s)
- Peng Cao
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Zejia Sun
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Chang Feng
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| | - Jiandong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Feilong Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Wei Wang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yong Zhao
- Transplantation Biology Research Division, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
21
|
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
|
22
|
Placental bed research: II. Functional and immunological investigations of the placental bed. Am J Obstet Gynecol 2019; 221:457-469. [PMID: 31288009 DOI: 10.1016/j.ajog.2019.07.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 06/01/2019] [Accepted: 07/02/2019] [Indexed: 01/14/2023]
Abstract
Research on the placenta as the interface between the mother and the fetus has been undertaken for some 150 years, and in 2 subsequent reviews, we attempted to summarize the situation. In the first part, we described the discovery of unique physiological modifications of the uteroplacental spiral arteries, enabling them to cope with a major increase in blood flow necessary to ensure proper growth of the fetus. These consist of an invasion of the arterial walls by trophoblast and a progressive disappearance of its normal structure. Researchers then turned to the pathophysiology of the placental bed and in particular to its maternal vascular tree. This yielded vital information for a better understanding of the so-called great obstetrical syndromes (preeclampsia, fetal growth restriction, premature labor and delivery, placenta accreta). Systematic morphological investigations of the uteroplacental vasculature showed that preeclampsia is associated with decreased or failed transformation of spiral arteries and the persistence of endothelial and smooth muscle cells in segments of their myometrial portion. Here we report on recent functional investigations of the placental bed, including in situ biophysical studies of uteroplacental blood flow and vascular resistance, and manipulation of uteroplacental perfusion. These new methodologies have provided a novel way of identifying pregnancies in which remodeling is impaired. In animals it is now possible to manipulate uteroplacental blood flow, leading to an enhancement of fetal growth; this opens the way to trials in abnormal human pregnancies. In this second part, we explored a new, extremely important area of research that deals with the role of specific subsets of leukocytes and macrophages in the placental bed. The human first-trimester decidua is rich in leukocytes called uterine natural killer cells. Both macrophages and uterine natural killer cells increase in number from the secretory endometrium to early pregnancy and play a critical role in mediating the process of spiral artery transformation by inducing initial structural changes. It seems therefore that vascular remodeling of spiral arteries is initiated independently of trophoblast invasion. Dysregulation of the immune system may lead to reproductive failure or pregnancy complications, and in this respect, recent studies have advanced our understanding of the mechanisms regulating immunological tolerance during pregnancy, with several mechanisms being proposed for the development of tolerance to the semiallogeneic fetus. In particular, these include several strategies by which the trophoblast avoids maternal recognition. Finally, an important new dimension is being explored: the likelihood that pregnancy syndromes and impaired uteroplacental vascular remodeling may be linked to future maternal and even the child's cardiovascular disease risk. The functional evidence underlying these observations will be discussed.
Collapse
|
23
|
Ahmadi M, Mohammadi M, Ali-Hassanzadeh M, Zare M, Gharesi-Fard B. MDSCs in pregnancy: Critical players for a balanced immune system at the feto-maternal interface. Cell Immunol 2019; 346:103990. [PMID: 31703912 DOI: 10.1016/j.cellimm.2019.103990] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/08/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) have emerged as a new immune regulator at the feto-maternal interface. Although the phenotypes and functions of these cells were primarily studied in pathological conditions such as cancers and infections, new evidence has underscored their beneficial roles in homeostasis and physiological circumstances such as normal pregnancy. In this regard, studies have shown an increased number of MDSCs, particularly granulocytic MDSCs, at the feto-maternal interface. These cells participate in maintaining immunological tolerance between mother and semi-allograft fetus through various mechanisms. They further seem to play critical roles in placentation and fetus growth process. The absence or dysregulation of MDSCs during pregnancy have been reported in several pregnancy complications. These cells are also abundant in the cord blood of neonates so as to balance the immune responses and prevent aggressive inflammatory responses. The current review summarizes and organizes detailed data on MDSCs and their roles during pregnancy.
Collapse
Affiliation(s)
- Moslem Ahmadi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mobin Mohammadi
- Cancer and Immunology Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran; Department of Immunology and Hematology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran; Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Zare
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Behrouz Gharesi-Fard
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Infertility Research Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
24
|
Ren J, Zeng W, Tian F, Zhang S, Wu F, Qin X, Zhang Y, Lin Y. Myeloid-derived suppressor cells depletion may cause pregnancy loss via upregulating the cytotoxicity of decidual natural killer cells. Am J Reprod Immunol 2019; 81:e13099. [PMID: 30737988 DOI: 10.1111/aji.13099] [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: 10/23/2018] [Revised: 01/12/2019] [Accepted: 01/28/2019] [Indexed: 12/21/2022] Open
Abstract
PROBLEM Maternal immune system tolerance to the semiallogeneic fetus is critical for a successful pregnancy. Studies have shown that myeloid-derived suppressor cells (MDSCs) play an important role in maintaining feto-maternal tolerance. However, the mechanisms remain poorly understood. METHODS Flow cytometry was used to evaluate the percentage of MDSCs in an allogeneic-normal-pregnant mouse model during different periods of gestation. We further assessed the percentage of MDSCs and their subtypes (granulocytic MDSCs [GR-MDSCs] and monocytic MDSCs [MO-MDSCs]) in a spontaneous abortion mouse model. The levels of the immunosuppressive molecules ARG-1, iNOS, IL-10, and TGF-β in MDSCs were also evaluated. MDSCs were depleted by anti-Gr-1 injection, and the resorption rate was calculated. The cytotoxicity of decidual natural killer (NK) cells was evaluated, and the percentage of regulatory NK (NKreg) cells and regulatory T lymphocytes (Tregs) were evaluated. RESULTS Myeloid-derived suppressor cells was accumulated in a time-dependent manner during pregnancy. However, the percentage of MDSCs was decreased in the spontaneous abortion mice compared with that in the control mice. In addition, the levels of ARG-1, iNOS, IL-10, and TGF-β in MDSCs decreased differentially. Finally, depletion of MDSCs was associated with increased rates of resorption and the proportion of NKreg and Treg cells in uterine tissues; meanwhile, the cytotoxicity of decidual NK cells was upregulated by increasing the level of perforin, granzyme B, and natural killer group protein 2 D-activating NK receptor (NKG2D). CONCLUSION Depletion of MDSCs may cause pregnancy loss, while upregulating the cytotoxicity of decidual NK cells and increasing NKreg and Treg cell numbers.
Collapse
Affiliation(s)
- Jiabin Ren
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Gynecologic Oncology, Shanghai, China
| | - Weihong Zeng
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fuju Tian
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Siming Zhang
- Department of Obstetrics and Gynecology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fan Wu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoli Qin
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Lin
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
25
|
Ren J, Zeng W, Tian F, Wu F, Zhang S, Liu X, Lin Y. Differential gene expression profile in monocytic myeloid-derived suppressor cells at maternal-fetal interface in a mouse model of spontaneous abortion. J Cell Physiol 2018; 234:10789-10799. [PMID: 30549043 DOI: 10.1002/jcp.27902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 10/23/2018] [Indexed: 12/25/2022]
Abstract
ABSTRACTBACKGROUND Monocytic myeloid-derived suppressor cells (MO-MDSCs) play an important role in maintaining normal pregnancy. However, it is still not clear what kind of changes in MO-MDSCs may lead to miscarriage, and which gene expression changes take place when MO-MDSCs migrate to the uterus as bone marrow-derived cells. METHODS We used flow sorting technology to obtain MO-MDSCs from the maternal-fetal interface and bone marrow, respectively. Affymetrix 3'IVT expression profiling chip technology was used to detect the differential gene expression profiles in MO-MDSCs at the maternal-fetal interface in a mouse model of spontaneous abortion compared with the normal fertility control mice. We also compared the differential gene expression of MO-MDSCs at the maternal-fetal interface compared with bone marrow in the normal fertility control mice. RESULTS We found that 3,409 genes in MO-MDSCs were upregulated and 1,539 genes were downregulated at the maternal-fetal interface in the spontaneous abortion mice compared with the normal fertility mice. These genes are enriched in cellular components, biological processes, molecular functions, and protein binding, tumor signaling pathway, the PI3K-Akt signaling pathway, intratumoral proteoglycans, and extracellular matrix receptor interactions. Furthermore, we found that 270 genes in MO-MDSCs were upregulated and 383 genes were downregulated at the maternal-fetal interface in the normal fertility mice compared with those in the bone marrow. These genes are enriched in cellular components, biological processes, molecular functions, cell cycle, tumor transcriptional disorder, and cell adhesion molecules. CONCLUSION Differential gene expression in MO-MDSCs likely contributes to a successful pregnancy in fetal-maternal immunotolerance.
Collapse
Affiliation(s)
- Jiabin Ren
- Department of Obstetrics and Gynecology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Key Laboratory of Gynecologic Oncology, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Weihong Zeng
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Fuju Tian
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Fan Wu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Siming Zhang
- Department of Obstetrics and Gynecology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Xiaorui Liu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Yi Lin
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
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
|
Salminen A, Kaarniranta K, Kauppinen A. Phytochemicals inhibit the immunosuppressive functions of myeloid-derived suppressor cells (MDSC): Impact on cancer and age-related chronic inflammatory disorders. Int Immunopharmacol 2018; 61:231-240. [DOI: 10.1016/j.intimp.2018.06.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/04/2018] [Accepted: 06/04/2018] [Indexed: 02/07/2023]
|
28
|
Koucký M, Malíčková K, Kopřivová H, Cindrová-Davies T, Hrbáčková H, Černý A, Šimják P, Pařízek A, Zima T. Low maternal serum concentrations of mannose-binding lectin are associated with the risk of shorter duration of pregnancy and lower birthweight. Scand J Immunol 2018; 88:e12675. [DOI: 10.1111/sji.12675] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/07/2018] [Indexed: 12/26/2022]
Affiliation(s)
- M. Koucký
- Department of Gynecology and Obstetrics of the First Faculty of Medicine and General Teaching Hospital; Charles University in Prague; Prague Czech Republic
| | - K. Malíčková
- Institute of Medical Biochemistry and Laboratory Diagnostics of the First Faculty of Medicine; General Teaching Hospital; Charles University in Prague; Prague Czech Republic
| | - H. Kopřivová
- Institute of Medical Biochemistry and Laboratory Diagnostics of the First Faculty of Medicine; General Teaching Hospital; Charles University in Prague; Prague Czech Republic
| | - T. Cindrová-Davies
- Department of Physiology, Development and Neuroscience; University of Cambridge; Cambridge UK
| | - H. Hrbáčková
- Department of Physiology, Development and Neuroscience; University of Cambridge; Cambridge UK
| | - A. Černý
- Department of Gynecology and Obstetrics of the First Faculty of Medicine and General Teaching Hospital; Charles University in Prague; Prague Czech Republic
| | - P. Šimják
- Department of Gynecology and Obstetrics of the First Faculty of Medicine and General Teaching Hospital; Charles University in Prague; Prague Czech Republic
| | - A. Pařízek
- Department of Gynecology and Obstetrics of the First Faculty of Medicine and General Teaching Hospital; Charles University in Prague; Prague Czech Republic
| | - T. Zima
- Institute of Medical Biochemistry and Laboratory Diagnostics of the First Faculty of Medicine; General Teaching Hospital; Charles University in Prague; Prague Czech Republic
| |
Collapse
|
29
|
Bronchud MH, Tresserra F, Xu W, Warren S, Cusido M, Zantop B, Zenclussen AC, Cesano A. Placental immune editing switch (PIES): learning about immunomodulatory pathways from a unique case report. Oncotarget 2018; 7:83817-83827. [PMID: 27852037 PMCID: PMC5347808 DOI: 10.18632/oncotarget.13306] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/25/2016] [Indexed: 12/22/2022] Open
Abstract
The hypothesis of this work is that, in order to escape the natural immune surveillance mechanisms, cancer cells and the surrounding microenvironment might express ectopically genes that are physiologically present in the placenta to mediate fetal immune-tolerance. These natural “placental immune-editing switch” mechanisms (PIES) may represent the result of millions of years of mammalian evolution developed to allow materno-fetal tolerance. Here, we introduce genes of the immune regulatory pathways that are either similarly over- or under-expressed in tumor vs normal tissue. Our analysis was carried out in primary breast cancer with metastatic homolateral axillary lymph nodes as well as placenta tissue (both uterine decidual tissue and term placenta tissue) from a pregnant woman. Gene expression profiling of paired non-self and self tissues (i.e. placenta/uterus; breast cancer/normal breast tissue; metastatic lymphnode/normal lymphnode tissue) was performed using the PanCancer Immune gene panel, a 770 Nanostring gene expression panel. Our findings reveal overlapping in specific immune gene expression in placenta and cancer tissue, suggesting that these genes might play an important role in maintaining immune tolerance both physiologically (in the placenta) and pathologically (in the cancer setting).
Collapse
Affiliation(s)
- Miguel H Bronchud
- Institut Bellmunt Oncologia, Hospital Universitario Dexeus. Grupo Quirón Salud, Barcelona, Spain
| | - Francesc Tresserra
- Servei Anatomia Patològica, Hospital Universitario Dexeus. Grupo Quirón Salud, Barcelona, Spain
| | - Wenjie Xu
- Nanostring Technologies, Immune Oncology, Seattle, WA, USA
| | - Sarah Warren
- Nanostring Technologies, Immune Oncology, Seattle, WA, USA
| | - Maite Cusido
- Ginecologia Oncològica, Hospital Universitario Dexeus. Grupo Quirón Salud, Barcelona, Spain
| | - Bernat Zantop
- Servei Obstetricia i Neonatologia, Hospital Universitario Dexeus. Grupo Quirón Salud, Barcelona, Spain
| | - Ana Claudia Zenclussen
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | | |
Collapse
|
30
|
Beaman KD, Jaiswal MK, Katara GK, Kulshreshta A, Pamarthy S, Ibrahim S, Kwak-Kim J, Gilman-Sachs A. Pregnancy is a model for tumors, not transplantation. Am J Reprod Immunol 2017; 76:3-7. [PMID: 27293114 DOI: 10.1111/aji.12524] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 04/27/2016] [Indexed: 02/06/2023] Open
Abstract
Nearly 65 years have passed since Peter Medawar posed the following question: "How does the pregnant mother contrive to nourish within itself, for many weeks or months, a fetus that is an antigenically foreign body." Now, understanding of reproductive immunology has demonstrated that the HLA antigens in the placenta are non-classical and do not induce rejection. In the placenta and in tumors, 50% or more of the cells are cells of the immune system and were once thought to be primed and ready for killing tumors or the "fetal transplant" but these cells are not potential killers but abet the growth of either the tumor or the placenta. We believe that these cells are there to create an environment, which enhances either placental or tumor growth. By examining the similarities of the placenta's and tumor's immune cells, novel mechanisms to cause tumors to be eliminated can be devised.
Collapse
Affiliation(s)
- Kenneth D Beaman
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Mukesh K Jaiswal
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Gajendra K Katara
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Arpita Kulshreshta
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Sahithi Pamarthy
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Safaa Ibrahim
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Joanne Kwak-Kim
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Alice Gilman-Sachs
- Clinical Immunology Laboratory, Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
31
|
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]
|
32
|
Lopez Gelston CA, Mitchell BM. Recent Advances in Immunity and Hypertension. Am J Hypertens 2017; 30:643-652. [PMID: 28200062 DOI: 10.1093/ajh/hpx011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/18/2017] [Indexed: 01/01/2023] Open
Abstract
Persistent immune system activation plays an important role in the development of various forms of hypertension. Activation of the innate immune system, inflammation, and subsequent adaptive immune system response causing end-organ injury and dysfunction ultimately leads to hypertension and its associated sequelae including coronary artery disease, heart failure, stroke, and chronic kidney disease. In this review, we will provide updates on the innate and adaptive immune cells involved in hypertension, the current understanding of how the immune system gets activated, and examine the recently discovered mechanisms involved in several forms of experimental hypertension.
Collapse
Affiliation(s)
- Catalina A Lopez Gelston
- Department of Medical Physiology, Texas A&M University Health Science Center, College Station, Texas, USA
| | - Brett M Mitchell
- Department of Medical Physiology, Texas A&M University Health Science Center, College Station, Texas, USA
| |
Collapse
|
33
|
Beaman KD, Dambaeva S, Katara GK, Kulshrestha A, Gilman-Sachs A. The immune response in pregnancy and in cancer is active and supportive of placental and tumor cell growth not their destruction. Gynecol Oncol 2017; 145:476-480. [PMID: 28477880 DOI: 10.1016/j.ygyno.2017.04.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/14/2017] [Accepted: 04/24/2017] [Indexed: 12/27/2022]
Abstract
While many investigators have described the biochemical and physiological similarities between tumor cells and trophoblast cells, in this discourse we will compare primarily their leucocytes, which constitute a large portion of the tumor and its microenvironment as well as the placenta and its microenvironment. There is a remarkable similarity between the cells that support placental growth and development and tumor growth and development. In many cases over half of the cells present in the tumor and the placenta are non-tumor or nontrophoblast cells, immune cells. Most of these immune cells are prevented from attacking the fetal derived placental cells and the self-derived tumor cells. Nevertheless, these leucocytes, in our opinion, are very active and support tumor and placental cell growth through the production of growth factors and angiogenic factors. These cells do this by activating the portion of the immune response which initiates and helps control tissue repair.
Collapse
Affiliation(s)
- Kenneth D Beaman
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| | - Svetlana Dambaeva
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Gajendra K Katara
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Arpita Kulshrestha
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| | - Alice Gilman-Sachs
- Department of Microbiology and Immunology, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
| |
Collapse
|
34
|
Ghaebi M, Nouri M, Ghasemzadeh A, Farzadi L, Jadidi-Niaragh F, Ahmadi M, Yousefi M. Immune regulatory network in successful pregnancy and reproductive failures. Biomed Pharmacother 2017; 88:61-73. [PMID: 28095355 DOI: 10.1016/j.biopha.2017.01.016] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 12/27/2016] [Accepted: 01/02/2017] [Indexed: 12/23/2022] Open
Abstract
Maternal immune system must tolerate semiallogenic fetus to establish and maintain a successful pregnancy. Despite the existence of several strategies of trophoblast to avoid recognition by maternal leukocytes, maternal immune system may react against paternal alloantigenes. Leukocytes are important components in decidua. Not only T helper (Th)1/Th2 balance, but also regulatory T (Treg) cells play an important role in pregnancy. Although the frequency of Tregs is elevated during normal pregnancies, their frequency and function are reduced in reproductive defects such as recurrent miscarriage and preeclampsia. Tregs are not the sole population of suppressive cells in the decidua. It has recently been shown that regulatory B10 (Breg) cells participate in pregnancy through secretion of IL-10 cytokine. Myeloid derived suppressor cells (MDSCs) are immature developing precursors of innate myeloid cells that are increased in pregnant women, implying their possible function in pregnancy. Natural killer T (NKT) cells are also detected in mouse and human decidua. They can also affect the fetomaternal tolerance. In this review, we will discuss on the role of different immune regulatory cells including Treg, γd T cell, Breg, MDSC, and NKT cells in pregnancy outcome.
Collapse
Affiliation(s)
- Mahnaz Ghaebi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- Department of Biochemistry and Clinical Laboratories, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aliyeh Ghasemzadeh
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Laya Farzadi
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
35
|
Kyvelidou C, Sotiriou D, Antonopoulou T, Tsagkaraki M, Tserevelakis GJ, Filippidis G, Athanassakis I. l-Carnitine affects preimplantation embryo development toward infertility in mice. Reproduction 2016; 152:283-91. [PMID: 27402869 DOI: 10.1530/rep-16-0290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 07/07/2016] [Indexed: 01/16/2023]
Abstract
l-Carnitine (l-Cn), despite the beneficial role as energy-generating substance delivering long-chain fatty acids to the β-oxidation pathway in mitochondria, has been accused to cause an endometriosis-like state to BALB/c mice manifested by increased inflammatory cytokines in serum and peritoneal fluid, accumulation of immune cells in the peritoneal cavity and uterine walls and most importantly, correlating to infertility. Exploring this type of infertility, the effect of l-Cn on preimplantation embryo development, ovarian integrity and systemic maternal immunity was studied. Using nonlinear microscopy analysis, which was shown to be a powerful tool for determining embryo quality by quantitatively estimating the lipid body (LB) content of the cells, it was shown that in vitro and in vivo administration of l-Cn significantly decreased LB mean area in zygotes. Daily intraperitoneal administration of 2.5mg l-Cn for 3, 4 and 7days to mice significantly decreased the percent of normal zygotes. However, only the 7-day treatment persisted by affecting 2- and 8-cell stage embryos, while almost abolishing blastocyst development. Such effects were accompanied by abnormal ovarian histology, showing increased numbers of corpora luteus and elevated progesterone concentration in the serum. In addition, it was shown that the 7-day l-Cn treatment pushed maternal systemic immunity toward inflammation and immunosuppression by increasing CD11b-, CD25- and CD11bGr1-positive cells in spleen, which opposed the necessity for immunostimulation at these early stages of pregnancy. In conclusion, the results presented here demonstrated that elevated doses of l-Cn affect early stages of embryo development, leading to infertility.
Collapse
Affiliation(s)
| | | | | | - Margarita Tsagkaraki
- Institute of Electronic Structure and LaserFoundation of Research and Technology, Crete, Greece Department of PhysicsUniversity of Crete, Crete, Greece
| | - George J Tserevelakis
- Institute of Electronic Structure and LaserFoundation of Research and Technology, Crete, Greece
| | - George Filippidis
- Institute of Electronic Structure and LaserFoundation of Research and Technology, Crete, Greece
| | | |
Collapse
|
36
|
Pan T, Liu Y, Zhong LM, Shi MH, Duan XB, Wu K, Yang Q, Liu C, Wei JY, Ma XR, Shi K, Zhang H, Zhou J. Myeloid-derived suppressor cells are essential for maintaining feto-maternal immunotolerance via STAT3 signaling in mice. J Leukoc Biol 2016; 100:499-511. [PMID: 27203698 DOI: 10.1189/jlb.1a1015-481rr] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/08/2016] [Indexed: 12/12/2022] Open
Abstract
Maternal immune system tolerance to the semiallogeneic fetus is essential for a successful pregnancy; however, the mechanisms underlying this immunotolerance have not been fully elucidated. Here, we demonstrate that myeloid-derived suppressor cells play an important role in maintaining feto-maternal tolerance. A significant expansion of granulocytic myeloid-derived suppressor cells was observed in multiple immune organs and decidual tissues from pregnant mice. Pregnancy-derived granulocytic myeloid-derived suppressor cells suppressed T cell responses in a reactive oxygen species-dependent manner and required direct cell-cell contact. Mechanistic studies showed that progesterone facilitated differentiation and activation of granulocytic myeloid-derived suppressor cells, mediated through STAT3 signaling. The STAT3 inhibitor JSI-124 and a specific short hairpin RNA completely abrogated the effects of progesterone on granulocytic myeloid-derived suppressor cells. More importantly, granulocytic myeloid-derived suppressor cell depletion dramatically enhanced the abortion rate in normal pregnant mice, whereas adoptive transfer of granulocytic myeloid-derived suppressor cells clearly reduced the abortion rate in the CBA/J X DBA/2J mouse model of spontaneous abortion. These observations collectively demonstrate that granulocytic myeloid-derived suppressor cells play an essential role in the maintenance of fetal immunotolerance in mice. Furthermore, our study supports the notion that in addition to their well-recognized roles under pathologic conditions, myeloid-derived suppressor cells perform important functions under certain physiologic circumstances.
Collapse
Affiliation(s)
- Ting Pan
- Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Yufeng Liu
- Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Li Mei Zhong
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Mao Hua Shi
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Xiao Bing Duan
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Kang Wu
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Qiong Yang
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Chao Liu
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Jian Yang Wei
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Xing Ru Ma
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China
| | - Kun Shi
- Department of Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Hui Zhang
- Institute of Human Virology, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Sun Yat-sen University, Guangzhou, China; and
| | - Jie Zhou
- Program in Immunology, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Institute of Human Virology, Sun Yat-sen University, Guangzhou, China; Key Laboratory of Tropical Disease Control, Chinese Ministry of Education, Sun Yat-sen University, Guangzhou, China; and
| |
Collapse
|