1
|
Mei J, Wu B, Li M, Ma L, Yang X, Ma Y, Huang Y. Effect of Cyclosporine A on Th1/Th2 Cytokine Production by Decidual Stromal Cells Mediated by Trophoblast-derived Galectin-9. Reprod Sci 2024; 31:1903-1914. [PMID: 38273122 DOI: 10.1007/s43032-023-01431-5] [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: 09/05/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024]
Abstract
This study aimed to investigate the effect of cyclosporine A (CsA) on secretion of Th1 and Th2 cytokines by decidual stromal cells (DSCs) mediated by galectin (Gal)-9.HTR8/SVneo cells and primary trophoblasts were used for in vitro studies. Gal-9 expression was measured using quantitative real-time polymerase chain reaction and enzyme-linked immunosorbent assay, CsA was used to regulate Gal-9 expression in trophoblasts. DSCs were treated with trophoblast supernatant and changes in Th1 and Th2 cytokine levels were analyzed. Changes in DSC levels of the T-cell immunoglobulin mucin receptor 3 (TIM-3) levels in DSCs after treatment with Gal-9 were assessed. Western blotting and ERK and AKT inhibitors were used to assess the involvement of the corresponding signaling pathways. Gal-9 was expressed by both primary trophoblasts and HTR8/SVneo cells. CsA treatment increased Gal-9 secretion by trophoblasts, which in turn increased IL-6 (Th2 cytokine) and decreased TNF-α and IFN-γ (Th1 cytokines) secretion in DSCs. Upon downregulation of trophoblast Gal-9 secretion, DSCs secreted lower levels of Th2 cytokines and higher levels of Th1 cytokines, and the effect was reversed by addition of CsA. TIM-3 expression changed in parallel with Gal-9 secretion. CsA treatment upregulated expression of Gal-9 in trophoblasts, promoted secretion of Th2 cytokines, and inhibited secretion of Th1 cytokines via ERK signaling.
Collapse
Affiliation(s)
- Jiaoqi Mei
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University) , Ministry of EducationThe First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- National Center for International Research "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, Haikou, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
| | - Bangyong Wu
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University) , Ministry of EducationThe First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- National Center for International Research "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, Haikou, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
| | - Mengyongwei Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University) , Ministry of EducationThe First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- National Center for International Research "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, Haikou, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
| | - Lina Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University) , Ministry of EducationThe First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- National Center for International Research "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, Haikou, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
| | - Xiaohui Yang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University) , Ministry of EducationThe First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- National Center for International Research "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, Haikou, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University) , Ministry of EducationThe First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China.
- Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.
- National Center for International Research "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, Haikou, China.
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China.
| | - Yuanhua Huang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University) , Ministry of EducationThe First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China.
- Department of Reproductive Medicine, Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China.
- National Center for International Research "China-Myanmar Joint Research Center for Prevention and Treatment of Regional Major Disease" By the Ministry of Science and Technology of China, Haikou, China.
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
2
|
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
|
3
|
Li QH, Zhao QY, Yang WJ, Jiang AF, Ren CE, Meng YH. Beyond Immune Balance: The Pivotal Role of Decidual Regulatory T Cells in Unexplained Recurrent Spontaneous Abortion. J Inflamm Res 2024; 17:2697-2710. [PMID: 38707955 PMCID: PMC11070170 DOI: 10.2147/jir.s459263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Recurrent spontaneous abortion (RSA) is defined as two or more consecutive pregnancy failures, which brings tremendous stress to women of childbearing age and seriously affects family well-being. However, the reason in about 50% of cases remains unknown and is defined as unexplained recurrent spontaneous abortion (URSA). The immunological perspective in URSA has attracted widespread attention in recent years. The embryo is regarded as a semi-allogeneic graft to the mother. A successful pregnancy requires transition to an immune environment conducive to embryo survival at the maternal-fetal interface. As an important member of regulatory immunity, regulatory T (Treg) cells play a key role in regulating immune tolerance at the maternal-fetal interface. This review will focus on the phenotypic plasticity and lineage stability of Treg cells to illustrate its relationship with URSA.
Collapse
Affiliation(s)
- Qing-Hui Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Qiu-Yan Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Wei-Jing Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Ai-Fang Jiang
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Chun-E Ren
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Yu-Han Meng
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| |
Collapse
|
4
|
Wiley KS, Kwon D, Knorr DA, Fox MM. Regulatory T-cell phenotypes in prenatal psychological distress. Brain Behav Immun 2024; 116:62-69. [PMID: 38016492 PMCID: PMC11402516 DOI: 10.1016/j.bbi.2023.11.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/18/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Experiencing symptoms of psychological distress during pregnancy is common and has been linked to dysregulated immune functioning. In this context, immunoregulatory function is especially relevant because of its crucial role in establishment and maintenance of healthy pregnancy. However, little research has examined associations between women's prenatal psychological distress and immunoregulatory biomarkers. We investigated how symptoms of depression, anxiety, and stress relate to circulating levels of regulatory T-cells (Tregs). MATERIALS AND METHODS Pregnant Latina women were assessed at around 12 weeks of pregnancy (N = 82). These assessments included blood draws and self-report questionnaires assessing symptoms of depression, state anxiety, pregnancy-related anxiety, and perceived stress. Flow cytometry on PBMCs was used to quantify circulating Tregs, defined as CD3+CD4+CD25hiCD127loFoxP3+, and subpopulations positive for one of the following intra- or extracellular markers, CD45RA, CTLA-4, Helios, PD-1, TIM-3, and TIGIT. We collected 82 samples at 12 weeks. Multivariable linear regressions tested for associations between symptoms of psychological distress and Treg concentrations, adjusted for gestational age. RESULTS State anxiety symptoms at 12 weeks were negatively associated with parent Treg cell levels (b = -4.02, p = 0.023) and subpopulations Helios+ (b = -3.29, p = 0.019) and TIM3+ (b = -3.17, p = 0.008). Perceived stress was negatively associated with the PD-1+ subpopulation at 12 weeks (b = -4.02, p = 0.023). Depression was not related to Tregs or the subpopulations. CONCLUSION Our observation that symptoms of anxiety and stress are related to tolerogenic immunology suggests a possible biomechanism explaining correlations of maternal mood disorders with adverse outcomes for mothers and offspring.
Collapse
Affiliation(s)
- Kyle S Wiley
- Department of Anthropology, University of California, Los Angeles, United States; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, United States.
| | - Dayoon Kwon
- Department of Epidemiology, UCLA Fielding School of Public Health, University of California, Los Angeles, United States
| | - Delaney A Knorr
- Department of Anthropology, University of California, Los Angeles, United States
| | - Molly M Fox
- Department of Anthropology, University of California, Los Angeles, United States; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, United States
| |
Collapse
|
5
|
Cui Y, Wu S, Liu K, Zhao H, Ma B, Gong L, Zhou Q, Li X. Extra villous trophoblast-derived PDL1 can ameliorate macrophage inflammation and promote immune adaptation associated with preeclampsia. J Reprod Immunol 2024; 161:104186. [PMID: 38134680 DOI: 10.1016/j.jri.2023.104186] [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: 08/31/2023] [Revised: 10/11/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION Severe preeclampsia (sPE) is a systemic syndrome that may originate from chronic inflammation. Maintaining maternal-fetal hemostasis by the co-inhibitory molecule programmed death ligand 1 (PDL1) can be favorable for ameliorating inflammation from immune cells. Apart from programmed death 1 (PD1) expression, decidual macrophages (dMs) produce inflammatory cytokines, in response to cells which express PDL1. However, strong evidence is lacking regarding whether the PDL1/PD1 interaction between trophoblasts and decidual macrophages affects inflammation during sPE development. METHODS To determine whether the trophoblast-macrophage crosstalk via the PDL1/PD1 axis modulates the inflammatory response in sPE-like conditions, at first, maternal-fetal tissues from sPE and normal patients were collected, and the PDL1/PD1 distribution was analyzed by Western blot, immunohistochemistry/ immunofluorescence and flow cytometry. Next, a coculture system was established and flow cytometry was used to identify how PDL1 was involved in macrophage-related inflammation under hypoxic stress. Transcriptional analysis was performed to clarify the inflammation-associated pathway induced by the PDL1/PD1 interaction. Finally, the Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME) mouse model was used to examine the effect of PDL1 on macrophage-related inflammation by measuring PE-like symptoms. RESULTS In maternal-fetal tissue from sPE patients, placental extravillous trophoblasts (EVTs) and dMs had a surprisingly increase of PDL1 and PD1 expression, respectively, accompanied by a higher percentage of CD68 +CD86 + dMs. In vitro experiments showed that trophoblast-derived PDL1 under hypoxia interacted with PD1 on CD14 +CD80 +macrophages, leading to suppression of inflammation through the TNFα-p38/NFκB pathway. Accordingly, the PE-like mouse model showed a reversal of PE-like symptoms and a reduced F4/80 + CD86 + macrophage percentage in the uterus in response to recombinant PDL1 protein administration, indicating the protective effect of PDL1. DISCUSSION Our results initially explained an immunological adaptation of trophoblasts under placental hypoxia, although this protection was insufficient. Our findings suggest the possible capacity of modulating PDL1 expression as a potential therapeutic strategy to target the inflammatory response in sPE.
Collapse
Affiliation(s)
- Yutong Cui
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Suwen Wu
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Ketong Liu
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Huanqiang Zhao
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China
| | - Bo Ma
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Lili Gong
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China
| | - Qiongjie Zhou
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China.
| | - Xiaotian Li
- Department Obstetrics, Obstetrics and Gynaecology Hospital of Fudan University, Shanghai, China; Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
6
|
Zych M, Roszczyk A, Dąbrowski F, Kniotek M, Zagożdżon R. Soluble Forms of Immune Checkpoints and Their Ligands as Potential Biomarkers in the Diagnosis of Recurrent Pregnancy Loss-A Preliminary Study. Int J Mol Sci 2023; 25:499. [PMID: 38203670 PMCID: PMC10779235 DOI: 10.3390/ijms25010499] [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: 10/17/2023] [Revised: 11/30/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Immune checkpoints (ICPs) serve as regulatory switches on immune-competent cells. Soluble ICPs consist of fragments derived from ICP molecules typically located on cell membranes. Research has demonstrated that they perform similar functions to their membrane-bound counterparts but are directly present in the bloodstream. Effective control of the maternal immune system is vital for a successful pregnancy due to genetic differences between the mother and fetus. Abnormalities in the immune response are widely acknowledged as the primary cause of spontaneous abortions. In our research, we introduce a novel approach to understanding the immune-mediated mechanisms underlying recurrent miscarriages and explore new possibilities for diagnosing and preventing pregnancy loss. The female participants in the study were divided into three groups: RSA (recurrent spontaneous abortion), pregnant, and non-pregnant women. The analysis of soluble forms of immune checkpoints and their ligands in the serum of the study groups was conducted using the Luminex method Statistically significant differences in the concentrations of (ICPs) were observed between physiological pregnancies and the RSA group. Among patients with RSA, we noted reduced concentrations of sGalectin-9, sTIM-3, and sCD155, along with elevated concentrations of LAG-3, sCD80, and sCD86 ICPs, in comparison to physiological pregnancies. Our study indicates that sGalectin-9, TIM-3, sLAG-3, sCD80, sCD86, sVISTA, sNectin-2, and sCD155 could potentially serve as biological markers of a healthy, physiological pregnancy. These findings suggest that changes in the concentrations of soluble immune checkpoints may have the potential to act as markers for early pregnancy loss.
Collapse
Affiliation(s)
- Michał Zych
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Mazovian Voivodeship, Poland; (M.Z.); (A.R.); (R.Z.)
| | - Aleksander Roszczyk
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Mazovian Voivodeship, Poland; (M.Z.); (A.R.); (R.Z.)
| | - Filip Dąbrowski
- Department of Obstetrics, Perinatology and Neonatology, Center of Postgraduate Medical Education, Ceglowska 80, 01-809 Warsaw, Mazovian Voivodeship, Poland;
- Club35, Polish Society of Obstetricians and Gynecologists PTGiP, Cybernetyki7F/87, 02-677 Warsaw, Mazovian Voivodeship, Poland
- 1st Department of Obstetrics and Gynecology, Medical University of Warsaw, Starynkiewicza 1, 02-015 Warsaw, Mazovian Voivodeship, Poland
| | - Monika Kniotek
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Mazovian Voivodeship, Poland; (M.Z.); (A.R.); (R.Z.)
| | - Radosław Zagożdżon
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Mazovian Voivodeship, Poland; (M.Z.); (A.R.); (R.Z.)
- Department of Immunology, Transplantology, and Internal Diseases, Medical University of Warsaw, Nowogrodzka 59, 02-006 Warsaw, Mazovian Voivodeship, Poland
| |
Collapse
|
7
|
Zhang Y, Wang H, Qiu P, Jiang J, Wu X, Mei J, Sun H. Decidual macrophages derived NO downregulates PD-L1 in trophoblasts leading to decreased Treg cells in recurrent miscarriage. Front Immunol 2023; 14:1180154. [PMID: 37520550 PMCID: PMC10379637 DOI: 10.3389/fimmu.2023.1180154] [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] [Received: 03/05/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Placental trophoblasts contribute to regulatory T (Treg) function via the programmed cell death-1 (PD-1)/PD-1 ligand 1 (PD-L1) pathway during normal pregnancy. Decreased expression of PD-L1 in trophoblasts was closely associated with Treg deficiency in the development of pregnancy failure. Thus, targeting PD-L1 might be a novel therapy to prevent pregnancy loss. However, the mechanisms for modulating the expression of PD-L1 in trophoblasts are an enigma. Methods The proportion of decidual Treg cells, and the profile of decidual macrophages (DMs) sampled from women with normal pregnancy (NP) and recurrent miscarriage (RM) were evaluated by flow cytometry. The expression of Yin and Yang 1 protein (YY1) and PD-L1 in human villous were measured by Immunohistochemistry (IHC), qRT-PCR and western blot. The determination of soluble PD-L1 (sPD-L1) in serum from NP and RM, and trophoblast conditioned media (TCM) was performed by the PD-L1 SimpleStep ELISA kit. Knockdown of YY1 was processed in the human trophoblast derived cell lines, HTR-8 and Bewo, with siYY1 transfection. Peripheral naïve CD4+ T cells were isolated from women with NP for the in vitro culture. The percentages of Treg cells differentiated from peripheral naïve CD4+ T cells were measured by flow cytometry. The interaction between YY1 and CD274 was proved by CHIP. The expression of inducible nitric oxide synthase (iNOS) in decidua was evaluated by IHC. The level of NO in serum from women with NP and RM was determined by the Griess reagent system. The effects of NO on YY1 were determined by the in vitro culture of HTR-8 cells with the NO donor, SNAP. The in vivo model comprising twelve pregnant mice and underwent different treatment. The percentages of Treg cells in murine uterus were measured by flow cytometry. Similarly, Western blot and IHC were performed to determine the expression of YY1 and PD-L1 in murine placenta. Results Decreased expression of YY1 and PD-L1 in trophoblasts and lower proportion of decidual Treg cells were observed in patients with RM. Knockdown of YY1 contributes to a lower expression of YY1 and PD-L1. Soluble PD-L1 in the supernatant from HTR-8 cells was also decreased with siYY1 administration. Lower Treg differentiation was observed in the presence of supernatant from HTR-8 cells treated with siYY1. CHIP analysis revealed that endogenous YY1 directly occupied the promoter region of the CD274 (PD-L1) gene. Accompanied with increased M1 DMs, higher NO was observed in serum sampled from patients with RM. In the presence of Reduced expression of YY1 and PD-L1 was observed in HTR-8 cells with the treatment of SNAP. Furthermore, less Treg differentiation was observed with SNAP treated TCM. Moreover, our in vivo data found that YY1 deficiency was associated with decreased PD-L1, which further resulting in less Treg differentiation and Treg deficiency at the maternal-fetal interface and increased embryo loss. Discussion Our work found the modulatory capacity of YY1 on PD-L1 in trophoblasts during early pregnancy. Furthermore, reduced YY1 was supposed resulting from higher levels of NO produced from the M1 DMs in RM.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Mei
- *Correspondence: Haixiang Sun, ; Jie Mei,
| | | |
Collapse
|
8
|
Zhang Y, Liu Z, Sun H. Fetal-maternal interactions during pregnancy: a 'three-in-one' perspective. Front Immunol 2023; 14:1198430. [PMID: 37350956 PMCID: PMC10282753 DOI: 10.3389/fimmu.2023.1198430] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023] Open
Abstract
A successful human pregnancy requires the maternal immune system to recognize and tolerate the semi-allogeneic fetus, allowing for appropriate trophoblasts invasion and protecting the fetus from invading pathogens. Therefore, maternal immunity is critical for the establishment and maintenance of pregnancy, especially at the maternal-fetal interface. Anatomically, the maternal-fetal interface has both maternally- and fetally- derived cells, including fetal originated trophoblasts and maternal derived immune cells and stromal cells. Besides, a commensal microbiota in the uterus was supposed to aid the unique immunity in pregnancy. The appropriate crosstalk between fetal derived and maternal originated cells and uterine microbiota are critical for normal pregnancy. Dysfunctional maternal-fetal interactions might be associated with the development of pregnancy complications. This review elaborates the latest knowledge on the interactions between trophoblasts and decidual immune cells, highlighting their critical roles in maternal-fetal tolerance and pregnancy development. We also characterize the role of commensal bacteria in promoting pregnancy progression. Furthermore, this review may provide new thought on future basic research and the development of clinical applications for pregnancy complications.
Collapse
Affiliation(s)
- Yonghong Zhang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhaozhao Liu
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
9
|
Chen RY, Zhu Y, Shen YY, Xu QY, Tang HY, Cui NX, Jiang L, Dai XM, Chen WQ, Lin Q, Li XZ. The role of PD-1 signaling in health and immune-related diseases. Front Immunol 2023; 14:1163633. [PMID: 37261359 PMCID: PMC10228652 DOI: 10.3389/fimmu.2023.1163633] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/02/2023] [Indexed: 06/02/2023] Open
Abstract
Programmed cell death 1 receptor (PD-1) and its ligands constitute an inhibitory pathway to mediate the mechanism of immune tolerance and provide immune homeostasis. Significantly, the binding partners of PD-1 and its associated ligands are diverse, which facilitates immunosuppression in cooperation with other immune checkpoint proteins. Accumulating evidence has demonstrated the important immunosuppressive role of the PD-1 axis in the tumor microenvironment and in autoimmune diseases. In addition, PD-1 blockades have been approved to treat various cancers, including solid tumors and hematological malignancies. Here, we provide a comprehensive review of the PD-1 pathway, focusing on the structure and expression of PD-1, programmed cell death 1 ligand 1 (PD-L1), and programmed cell death 1 ligand 2 (PD-L2); the diverse biological functions of PD-1 signaling in health and immune-related diseases (including tumor immunity, autoimmunity, infectious immunity, transplantation immunity, allergy and immune privilege); and immune-related adverse events related to PD-1 and PD-L1 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Qiang Lin
- *Correspondence: Qiang Lin, ; Xiao-Zhong Li,
| | | |
Collapse
|
10
|
Chen Z, Huang J, Kwak-Kim J, Wang W. Immune checkpoint inhibitors and reproductive failures. J Reprod Immunol 2023; 156:103799. [PMID: 36724630 DOI: 10.1016/j.jri.2023.103799] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 01/02/2023] [Accepted: 01/06/2023] [Indexed: 01/15/2023]
Abstract
The human conceptus is a semi-allograft, which is antigenically foreign to the mother. Hence, the implantation process needs mechanisms to prevent allograft rejection during successful pregnancy. Immune checkpoints are a group of inhibitory pathways expressed on the surface of various immune cells in the form of ligand receptors. Immune cells possess these pathways to regulate the magnitude of immune responses and induce maternal-fetal tolerance. Briefly, 1) CTLA-4 can weaken T cell receptor (TCR) signals and inhibit T cell response; 2) The PD-1/PD-L1 pathway can reduce T cell proliferation, enhance T cell anergy and fatigue, reduce cytokine production, and increase T regulatory cell activity to complete the immunosuppression; 3) TIM3 interacts with T cells by binding Gal-9, weakening Th1 cell-mediated immunity and T cell apoptosis; 4) The LAG-3 binding to MHC II can inhibit T cell activation by interfering with the binding of CD4 to MHC II, and; 5) TIGIT can release inhibitory signals to NK and T cells through the ITIM structure of its cytoplasmic tail. Therefore, dysregulated immune checkpoints or the application of immune checkpoint inhibitors may impair human reproduction. This review intends to deliver a comprehensive overview of immune checkpoints in pregnancy, including CTLA-4, PD-1/PD-L1, TIM-3, LAG-3, TIGIT, and their inhibitors, reviewing their roles in normal and pathological human pregnancies.
Collapse
Affiliation(s)
- Zeyang Chen
- School of Medicine, Qingdao University, 38 Dengzhou Road, Qingdao 266000, PR China; Reproduction Medical Center, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China
| | - Jinxia Huang
- Reproduction Medical Center, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China; Department of Gynecology, Weihai Central Hospital Affiliated to Qingdao University, 3 Mishan East Road, Weihai 264400, PR China
| | - Joanne Kwak-Kim
- Reproductive Medicine and Immunology, Obstetrics and Gynecology, Clinical Sciences Department, Chicago Medical School, Rosalind Franklin University of Medicine and Science, Vernon Hills, IL 60061, USA; Center for Cancer Cell Biology, Immunology and Infection, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA.
| | - Wenjuan Wang
- Reproduction Medical Center, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China.
| |
Collapse
|
11
|
Hou R, Huang R, Zhou Y, Lin D, Xu J, Yang L, Wei X, Xie Z, Zhou Q. Single-cell profiling of the microenvironment in decidual tissue from women with missed abortions. Fertil Steril 2023; 119:492-503. [PMID: 36528108 DOI: 10.1016/j.fertnstert.2022.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 12/04/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To define the decidual microenvironment in euploid and aneuploid missed abortions and elective termination of pregnancies. DESIGN Prospective, multicenter, observational study. SETTING Tertiary hospital and descriptive analysis of transcriptomic data. PATIENT(S) A total of 34 patients experienced abortions, including 6 women who underwent elective terminations of pregnancy of unplanned pregnancies and 28 cases with missed abortions. All patients underwent their operations from Sep, 2021 to Sep, 2022. INTERVENTION(S) All women underwent villous copy number variation sequencing. Meanwhile, single-cell RNA sequencing were performed in the decidual tissues of 16 women, and reverse transcription quantitative polymerase chain reaction were performed in the decidual tissues of 18 women. MAIN OUTCOME MEASURE(S) Single-cell RNA sequencing was used to explore the changes in the microenvironment of decidual tissues in abortions. RESULT(S) Single-cell RNA sequencing indicated that the microenvironment of the decidual tissue of the missed-abortion group was altered, and that the stromal cells (SCs), natural killer cells, macrophages, and epithelial cells all reflected functional imbalances compared with the elective terminations of pregnancy group. We also noted a correlation between the proportion of senescent SCs and chromosomal abnormalities in missed-abortion embryos. The proportion of senescent decidual SCs in the decidual tissue of missed-abortion patients with common chromosomal abnormalities of the fetus was higher, and this was not conducive to fetal growth and was closely related to missed abortion. In addition, we ascertained that the strength of the HLA-KIR interaction between NK1 and NK2 subsets and non-senescent stromal cell subsets in the missed abortion decidual tissues was weakened, potentially playing a role in the occurrence of missed abortion. CONCLUSION(S) The decidualization of SCs in the missed-abortion decidual tissues was impaired, the clearance of senescent SCs by NK cells was weakened, the killing toxicity of non-senescent SCs was enhanced, macrophages were insufficiently resident at the maternal-fetal interface, and epithelial cell differentiation was unbalanced-all creating a maternal microenvironment that was not conducive to fetal growth. We posit that interfering with the expression of dysregulated genes in the missed-abortion decidual tissues and reversing the maternal microenvironment might constitute an effective means toward improving the clinical outcome of missed abortions. Intriguingly, we observed a correlation between stromal cell senescence and embryonic chromosomal abnormalities. Thus, we hypothesize that the DIO2 marker of senescent SCs can be used as a risk indicator for the occurrence of missed miscarriages with chromosomal abnormalities of the embryos, and that it can be applied to guide the clinical diagnosis and treatment of recurrent abortion. CLINICAL TRIAL REGISTRATION NUMBER NCT04425317.
Collapse
Affiliation(s)
- Ren Hou
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, People's Republic of China; Department of Eugenics and Genetics, The Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Renliang Huang
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, People's Republic of China
| | - Yanling Zhou
- Department of Eugenics and Genetics, The Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Dan Lin
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, People's Republic of China
| | - Jing Xu
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, People's Republic of China
| | - Liuqing Yang
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, People's Republic of China
| | - Xiaolan Wei
- Beijing SeekGene BioSciences, Beijing, People's Republic of China
| | - Zhuoming Xie
- Beijing SeekGene BioSciences, Beijing, People's Republic of China
| | - Qiaomiao Zhou
- Department of Genetics and Prenatal Diagnosis, Hainan Women and Children's Medical Center, Haikou, People's Republic of China.
| |
Collapse
|
12
|
Duan Y, Liu Y, Xu Y, Zhou C. Bioinformatics Analysis Identifies Key Genes in Recurrent Implantation Failure Based on Immune Infiltration. Reprod Sci 2023; 30:952-965. [PMID: 36045247 DOI: 10.1007/s43032-022-01060-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/04/2022] [Indexed: 11/25/2022]
Abstract
Recurrent implantation failure (RIF) is a thorny problem often encountered in the field of assisted reproduction. Existing evidences suggest that immune dysregulation may be involved in the pathogenesis of RIF. The purpose of this study is to explore immune-related genes contributing to RIF through data mining. The endometrial expression profiles of 24 RIF and 24 controls were obtained from the GEO database. The immune infiltration in bulk tissue was estimated by single sample gene set enrichment analysis (ssGSEA) method based on marker gene sets for immune cells generated from endometrial single-cell RNA sequencing data. The results showed that the infiltration levels of B cells and regulatory T cells (Tregs) were significantly reduced in the RIF group. Four hub genes (GJA1, PRKAG2, CPT1A, and ICA1) were identified by integrated analysis of weighted gene co-expression network analysis (WGCNA), random forest and LASSO regression. Moreover, these hub genes were significantly correlated with certain immune-related factors, especially CXCL12, CEACAM1, and XCR1. Single-gene GSEA indicated that the pathways associated with hub genes included the regulation of cell cycle, the process of epithelial-mesenchymal transition and transplant rejection, etc. A predictive model for RIF was constructed based on hub genes and performed well in the training dataset and the other two external datasets. Thus, this study identified immune-related key genes in RIF and provided new biomarkers for early diagnosis.
Collapse
Affiliation(s)
- Yuwei Duan
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, 510080, Guangdong, China
| | - Yongxiang Liu
- Department of Reproductive Medicine, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Yanwen Xu
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, 510080, Guangdong, China
| | - Canquan Zhou
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, Guangdong, China.
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
13
|
Wang S, Liu Y, Liang Y, Sun L, Du X, Shi Y, Meng J. Excessive Immune Activation and the Correlation with Decreased Expression of PD-1 at the Maternal-Fetal Interface in Preeclampsia. Reprod Sci 2023; 30:192-202. [PMID: 35708884 DOI: 10.1007/s43032-022-01003-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 06/07/2022] [Indexed: 01/11/2023]
Abstract
The etiology of preeclampsia (PE) is still unknown, and excessive immune activation is an important component of its pathogenesis. Programmed cell death protein 1 (PD-1) is one of immune checkpoints which may prevent overactivated immune attack and lead to a tolerant immune microenvironment. Little is known about the involvement of PD-1-mediated immunoregulation at the maternal-fetal interface in PE. To investigate the inflammatory pattern and the involvement of PD-1 in the decidua of women with PE, decidual tissues were obtained from PE and control pregnant women. Quantitative RT-PCR analysis of the mRNA levels of the inflammatory cytokines was performed. PD-1 expression was detected by immunohistochemistry, western blot analysis, and flow cytometry. To prove the role of PD-1, decidual immune cells were incubated with blocking antibodies, and the inflammatory cytokines were detected by ELISA. We observed that the mRNA levels of IL-1β, IL-6, TNF-α, and IFN-γ were higher in the decidua of the PE group than in the decidua of the control group. The mRNA levels of IL-4 and IL-10 were lower in PE. The expression level of PD-1 was significantly downregulated, and the proportion (%) of PD-1 + CD45 + cells was significantly lower in PE. There was a significant linear correlation between PD-1 expression and common proinflammatory cytokines in the decidua. Anti-PD-1 blocking antibody significantly increased the secretion of proinflammatory cytokines. Our data suggested that the inflammatory pattern and decreased PD-1 expression in the decidua might play an active role in the local immunoregulatory mechanisms of PE. The PD-1 pathway in the maternal-fetal interface possibly function to break the tolerant immune microenvironment in PE via inflammatory cytokines.
Collapse
Affiliation(s)
- Shan Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, Jinan, 250021, Shandong Province, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yining Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yue Liang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Lina Sun
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaoxiao Du
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, Jinan, 250021, Shandong Province, China
| | - Yueyang Shi
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Jinlai Meng
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, Jinan, 250021, Shandong Province, China.
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
- Second Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
- Key Laboratory of Birth Regulation and Control Technology of National Health and Family Planning Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, Shandong, China.
| |
Collapse
|
14
|
Effect of B7-H4 downregulation induced by Toxoplasma gondii infection on dysfunction of decidual macrophages contributes to adverse pregnancy outcomes. Parasit Vectors 2022; 15:464. [PMID: 36514159 PMCID: PMC9746109 DOI: 10.1186/s13071-022-05560-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/20/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Toxoplasma gondii infection during pregnancy can lead to fetal defect(s) or congenital complications. The inhibitory molecule B7-H4 expressed on decidual macrophages (dMφ) plays an important role in maternal-fetal tolerance. However, the effect of B7-H4 on the function of dMφ during T. gondii infection remains unclear. METHODS Changes in B7-H4 expression on dMφ after T. gondii infection were explored both in vivo and in vitro. B7-H4-/- pregnant mice (pregnant mice with B7-H4 gene knockout) and purified primary human dMφ treated with B7-H4 neutralizing antibody were used to explore the role of B7-H4 signaling on regulating the membrane molecules, synthesis of arginine metabolic enzymes and cytokine production by dMφ with T. gondii infection. Also, adoptive transfer of dMφ from wild-type (WT) pregnant mice or B7-H4-/- pregnant mice to infected B7-H4-/- pregnant mice was used to examine the effect of B7-H4 on adverse pregnancy outcomes induced by T. gondii infection. RESULTS The results illustrated that B7-H4-/- pregnant mice infected by T. gondii had poorer pregnancy outcomes than their wild-type counterparts. The expression of B7-H4 on dMφ significantly decreased after T. gondii infection, which resulted in the polarization of dMφ from the M2 toward the M1 phenotype by changing the expression of membrane molecules (CD80, CD86, CD163, CD206), synthesis of arginine metabolic enzymes (Arg-1, iNOS) and production of cytokines (IL-10, TNF-α) production. Also, we found that the B7-H4 downregulation after T. gondii infection increased iNOS and TNF-α expression mediated through the JAK2/STAT1 signaling pathway. In addition, adoptive transfer of dMφ from a WT pregnant mouse donor rather than from a B7-H4-/- pregnant mouse donor was able to improve adverse pregnancy outcomes induced by T. gondii infection. CONCLUSIONS The results demonstrated that the downregulation of B7-H4 induced by T. gondii infection led to the dysfunction of decidual macrophages and contributed to abnormal pregnancy outcomes. Moreover, adoptive transfer of B7-H4+ dMφ could improve adverse pregnancy outcomes induced by T. gondii infection.
Collapse
|
15
|
Guo F, Huang Y, Fernando T, Shi Y. Altered Molecular Pathways and Biomarkers of Endometrial Receptivity in Infertile Women with Polycystic Ovary Syndrome. Reprod Sci 2022; 29:3335-3345. [PMID: 35006579 DOI: 10.1007/s43032-022-00845-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 12/31/2021] [Indexed: 12/14/2022]
Abstract
Anovulation is the most prominent cause of infertility in polycystic ovary syndrome (PCOS) patients. Although ovulation can be corrected pharmacologically, the number of pregnancies remains low. Even if excellent embryos are transferred by IVF, it does not change the high miscarriage rate of PCOS patients. These facts collectively indicate that there is a disorder of endometrial development and receptivity to the embryo in PCOS patients, including the decrease of receptive ability, inhibition of embryo adhesion, undersupply of energy, poor blood perfusion, and pro-inflammatory status in the endometrium. However, it has never received the same attention as ovulatory dysfunction. Here we list some alternations of endometrial receptivity in women with PCOS, discuss the underlying intricate mechanisms, and try to find out the possible therapeutic targets, which may bring new perspectives to those who are able to provide high-quality embryos.
Collapse
Affiliation(s)
- Fei Guo
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Yufan Huang
- Department of Pharmacy, Mindong Hospital, Fujian Medical University, Ningde, 355000, Fujian, China
| | - Taniya Fernando
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China
| | - Yingli Shi
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
16
|
Kim J, Tran ANT, Lee JY, Park SH, Park SR, Min BH, Choi BH. Human Fetal Cartilage-Derived Progenitor Cells Exhibit Anti-Inflammatory Effect on IL-1β-Mediated Osteoarthritis Phenotypes In Vitro. Tissue Eng Regen Med 2022; 19:1237-1250. [PMID: 35932427 PMCID: PMC9679083 DOI: 10.1007/s13770-022-00478-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/18/2022] [Accepted: 06/27/2022] [Indexed: 10/15/2022] Open
Abstract
BACKGROUND In this study, we have investigated whether human fetal cartilage progenitor cells (hFCPCs) have anti-inflammatory activity and can alleviate osteoarthritis (OA) phenotypes in vitro. METHODS hFCPCs were stimulated with various cytokines and their combinations and expression of paracrine factors was examined to find an optimal priming factor. Human chondrocytes or SW982 synoviocytes were treated with interleukin-1β (IL-1β) to produce OA phenotype, and co-cultured with polyinosinic-polycytidylic acid (poly(I-C))-primed hFCPCs to address their anti-inflammatory effect by measuring the expression of OA-related genes. The effect of poly(I-C) on the surface marker expression and differentiation of hFCPCs into 3 mesodermal lineages was also examined. RESULTS Among the priming factors tested, poly(I-C) (1 µg/mL) most significantly induced the expression of paracrine factors such as indoleamine 2,3-dioxygenase, histocompatibility antigen, class I, G, tumor necrosis factor- stimulated gene-6, leukemia inhibitory factor, transforming growth factor-β1 and hepatocyte growth factor from hFCPCs. In the OA model in vitro, co-treatment of poly(I-C)-primed hFCPCs significantly alleviated IL-1β-induced expression of inflammatory factors such as IL-6, monocyte chemoattractant protein-1 and IL-1β, and matrix metalloproteinases in SW982, while it increased the expression of cartilage extracellular matrix such as aggrecan and collagen type II in human chondrocytes. We also found that treatment of poly(I-C) did not cause significant changes in the surface marker profile of hFCPCs, while showed some changes in the 3 lineages differentiation. CONCLUSION These results suggest that poly(I-C)-primed hFCPCs have an ability to modulate inflammatory response and OA phenotypes in vitro and encourage further studies to apply them in animal models of OA in the future.
Collapse
Affiliation(s)
- Jiyoung Kim
- Department of Physiology and Biophysics, Inha University College of Medicine, Incheon, 22212, Korea
| | - An Nguyen-Thuy Tran
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea.,Cell Therapy Center, Ajou University Medical Center, Suwon, 16499, Korea
| | - Ji Young Lee
- Department of Biomedical Sciences, Inha University College of Medicine, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Korea
| | - Sang-Hyug Park
- Department of Biomedical Engineering, Pukyong National University, Pusan, 48513, Korea
| | - So Ra Park
- Department of Physiology and Biophysics, Inha University College of Medicine, Incheon, 22212, Korea
| | - Byoung-Hyun Min
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Korea.,Cell Therapy Center, Ajou University Medical Center, Suwon, 16499, Korea
| | - Byung Hyune Choi
- Department of Biomedical Sciences, Inha University College of Medicine, 100 Inha-ro, Michuhol-gu, Incheon, 22212, Korea.
| |
Collapse
|
17
|
Zha Y, Liu H, Lin X, Yu L, Gao P, Li Y, Wu M, Gong X, Bian X, Kang Q, Zhi P, Dang X, Wang J, Feng L, Qiao F, Huang Y, Zeng W. Immune Deviation in the Decidua During Term and Preterm Labor. Front Immunol 2022; 13:877314. [PMID: 35757768 PMCID: PMC9226582 DOI: 10.3389/fimmu.2022.877314] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
The maternal-fetal immune disorder is considered to be an important factor of preterm birth (PTB); however, the underlying mechanism is still not fully understood. This study was designed to explore the innate and adaptive immune features in the decidua during term and preterm labor. Women delivered at term or preterm were classified into four groups: term not in labor (TNL, N=19), term in labor (TL, N=17), preterm not in labor (PNL, N=10), and preterm in labor (PIL, N=10). Decidua basalis and parietalis were collected and analyzed for macrophage subtypes (M1 and M2) as well as T helper 1 (Th1), Th2, Th17 and regulatory T (Treg) cells by flow cytometry and immunohistochemistry. Our results demonstrated significantly decreased frequencies of M2 cells and elevated M1/M2 ratio in the PIL group compared to that in the PNL group in both decidua basalis and parietalis, whereas no significant differences were found between the above two groups in both sites in terms of the polarization status of Th cells. On the contrary, macrophage subsets were comparable in the TL and TNL groups, whereas elevated Th1 percentages and Th1/Th2 ratio were observed in TL women compared to that in TNL women in the decidua. Interestingly, although the frequencies and ratios of Th17 and Treg were comparable among the four groups, the Th17/Treg ratios of these groups were significantly increased in decidua basalis than that in decidua parietalis. Collectively, the M1/M2 imbalance is associated with the breakdown of maternal-fetal immune tolerance during PTB, whereas the aberrant Th1/Th2 profile plays an important role in immune disorder during term labor. Moreover, Th17/Treg deviation is more remarkable in decidua basalis than in decidua parietalis.
Collapse
Affiliation(s)
- Ying Zha
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haiyi Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingguang Lin
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long Yu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqi Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xun Gong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyi Bian
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Kang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Zhi
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohe Dang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fuyuan Qiao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wanjiang Zeng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Zhao SJ, Muyayalo KP, Luo J, Huang D, Mor G, Liao AH. Next generation of immune checkpoint molecules in maternal-fetal immunity. Immunol Rev 2022; 308:40-54. [PMID: 35234305 DOI: 10.1111/imr.13073] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/09/2022] [Indexed: 12/18/2022]
Abstract
Successful pregnancy is a unique situation requires the maternal immune system to recognize and tolerate a semi-identical fetus and allow normal invasion of trophoblast cells. Although efforts have been made, the deep mechanisms of the maternal-fetal crosstalk have not yet been fully deciphered. Immune checkpoint molecules (ICMs) are a group of negative modulators of the immune response that avoid immune damage. They have been extensively studied in the fields of oncology and transplantation, while the latest evidence suggests that they are closely associated with pregnancy outcomes via multiple inhibitory mechanisms. Although studies have mostly demonstrated the regulatory role of the well-known PD-1, CTLA-4 at the maternal-fetal interface, what is unique about the newly discovered multiple ICMs remains a mystery. Here, we review the latest knowledge on ICMs, focusing on the first generation of checkpoints (PD-1, CTLA-4) and the next generation (Tim-3, Tigit, Lag-3, VISTA) highlighting their immunoregulatory roles in maternal-fetal tolerance and decidual vascular remodeling, and their involvement in pathological pregnancies. The content covers three aspects: the characteristics they possess, the dynamic expression profile of their expression at the maternal-fetal interface, and their involvement in pathological pregnancy. In immunotherapy strategies for pregnancy complications, upregulation of immune checkpoints may play a role. Meanwhile, the impact on pregnancy outcomes when using ICMs in clinical cancer treatment during pregnancy is a topic worth exploring. These may serve as a guide for future basic research and clinical applications of maternal-fetal immunity.
Collapse
Affiliation(s)
- Si-Jia Zhao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kahindo P Muyayalo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Donghui Huang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gil Mor
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, Michigan, USA
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Hu J, Qin X, Zhang J, Zhu Y, Zeng W, Lin Y, Liu X. Polystyrene microplastics disturb maternal-fetal immune balance and cause reproductive toxicity in pregnant mice. Reprod Toxicol 2021; 106:42-50. [PMID: 34626775 DOI: 10.1016/j.reprotox.2021.10.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/14/2021] [Accepted: 10/04/2021] [Indexed: 12/11/2022]
Abstract
Microplastics (MPs), which are emerging as a new type of environmental pollutants, have raised great concerns regarding their threats to human health. A successful pregnancy depends on the sophisticated regulation of the maternal-fetal immune balance, but the risks of polystyrene MP (PS-MP) exposure in early pregnancy remain unclear. In this study, we exposed the C57BL/6-mated BALB/c mice to PS-MP particles and used the flow cytometry to explore threats towards the immune system. Herein, the allogeneic mating murine model showed an elevated embryo resorption rate with a 10 μm PS-MP particle exposure during the peri-implantation period. Both the number and diameter of uterine arterioles decreased, which might reduce the uterine blood supply. Moreover, the percentage of decidual natural killer cells was reduced, whereas the helper T cells in the placenta increased. In addition, the M1/M2 ratio in macrophages reversed significantly to a dominant M2-subtype. Lastly, the cytokine secretion shifted towards an immunosuppressive state. Overall, our results demonstrated that PS-MPs have the potential to cause adverse effects on pregnancy outcomes via immune disturbance, providing new insights into the study of reproductive toxicity of MP particles in the human body.
Collapse
Affiliation(s)
- Jianing Hu
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xiaoli Qin
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Jinwen Zhang
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yueyue Zhu
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Weihong Zeng
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yi Lin
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Xiaorui Liu
- Institute of Shanghai Key Laboratory of Embryo Original Diseases and Shanghai Municipal Key Clinical Specialty Project Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China; The International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
20
|
Yang D, Dai F, Yuan M, Zheng Y, Liu S, Deng Z, Tan W, Chen L, Zhang Q, Zhao X, Cheng Y. Role of Transforming Growth Factor-β1 in Regulating Fetal-Maternal Immune Tolerance in Normal and Pathological Pregnancy. Front Immunol 2021; 12:689181. [PMID: 34531852 PMCID: PMC8438197 DOI: 10.3389/fimmu.2021.689181] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is composed of three isoforms, TGF-β1, TGF-β2, and TGF-β3. TGF-β1 is a cytokine with multiple biological functions that has been studied extensively. It plays an important role in regulating the differentiation of immune cells and maintaining immune cell functions and immune homeostasis. Pregnancy is a carefully regulated process. Controlled invasion of trophoblasts, precise coordination of immune cells and cytokines, and crosstalk between trophoblasts and immune cells play vital roles in the establishment and maintenance of normal pregnancy. In this systematic review, we summarize the role of TGF-β1 in regulating fetal-maternal immune tolerance in healthy and pathological pregnancies. During healthy pregnancy, TGF-β1 induces the production of regulatory T cells (Tregs), maintains the immunosuppressive function of Tregs, mediates the balance of M1/M2 macrophages, and regulates the function of NK cells, thus participating in maintaining fetal-maternal immune tolerance. In addition, some studies have shown that TGF-β1 is dysregulated in patients with recurrent spontaneous abortion or preeclampsia. TGF-β1 may play a role in the occurrence and development of these diseases and may be a potential target for the treatment of these diseases.
Collapse
Affiliation(s)
- Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengqin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yajing Zheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Tan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liping Chen
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianjie Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaomiao Zhao
- Department of Obstetrics and Gynecology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
21
|
Differences in Immune Checkpoints Expression (TIM-3 and PD-1) on T Cells in Women with Recurrent Miscarriages-Preliminary Studies. J Clin Med 2021; 10:jcm10184182. [PMID: 34575293 PMCID: PMC8468868 DOI: 10.3390/jcm10184182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Immune checkpoints are molecules that regulate the function of immune cells and control inflammation processes. An important role in this regard is played by TIM-3/Gal-9 and PD-1/PDL-1 interactions. Previous research performed in a mouse model of pregnancy loss confirmed that blocking TIM-3 could induce fetal loss. Similarly, the PD-1 molecule maintains protective interactions between the mother’s immune cells and the fetus. The purpose of this study was to assess the expression of these molecules on a range of T lymphocyte subpopulations from non-pregnant women with recurrent spontaneous abortion (RSA) versus healthy fertile women. Methods: PBMCs were isolated by gradient centrifugation of blood obtained from 12 healthy women and 24 women with RSA and immediately stained for flow cytometry analysis. Standard immunophenotyping of PBMC was performed with the antibodies against classical lymphocyte markers: CD3, CD4, CD8, and CD56. Immune checkpoints were investigated using antibodies against PD-1(CD279) and TIM-3(CD366). Results: We found that expression of TIM-3 was significantly decreased on CD8+ T lymphocytes in the RSA group, and expression of PD-1 was upregulated on CD4+ T lymphocytes in the RSA group in comparison to the healthy controls. Conclusions: Considering our findings, therapeutic intervention towards immune checkpoints may be a promising treatment option for recurrent spontaneous abortion.
Collapse
|
22
|
Wang S, Chen C, Sun F, Li M, Du M, Li X, Zhang Y. Involvement of the Tim-3 Pathway in the Pathogenesis of Pre-Eclampsia. Reprod Sci 2021; 28:3331-3340. [PMID: 34231168 DOI: 10.1007/s43032-021-00675-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/17/2021] [Indexed: 01/06/2023]
Abstract
Current methods of early diagnosis and prevention of pre-eclampsia (PE) are limited; the only available definite treatment is the initiation of delivery and complete removal of the placenta. Inappropriate activation of the immune system is thought to play considerable roles in PE. T cell immunoglobulin mucin-3 (Tim-3) has been reported to regulate immune responses and play important roles in maternal-fetal tolerance during early pregnancy. In this study, we investigated the functional regulation of Tim-3 in the maternal-fetal crosstalk during 3rd-trimester healthy pregnancy and its possible role in the pathogenesis of PE. We found that Tim-3 expression on decidual immune cells was associated with production of anti-inflammatory cytokines. Tim-3 pathway blockade resulted in higher IFN-γ but lower IL-4 and IL-10 production. Using a tube formation assay between HTR8/SVneo cells and human umbilical vein endothelial cells, we found that Tim-3 pathway blockade inhibits tube formation and reversed by addition of recombinant IL-4 and/or IL-10. Pre-eclamptic patients showed reduced Tim-3 expression on both decidual and peripheral immune cells (especially on peripheral CD8+T cells). Therefore, we proposed that abnormal Tim-3 signal resulted in immunological imbalance at the maternal-fetal interface and may be involved in the progress of PE by affecting uterine spiral artery remodeling. Our study expanded the regulatory function of Tim-3 signaling pathway to the 3rd-trimester pregnancy and provided a new target for early warning and therapeutic strategies of PE.
Collapse
Affiliation(s)
- Songcun Wang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Chunqin Chen
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Fengrun Sun
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Mengdie Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Meirong Du
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Xiaotian Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China. .,The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China. .,Institutes of Biochemical Sciences, Fudan University, Shanghai, China. .,The Shanghai Key Laboratory of Birth Defects, Shanghai, China.
| | - Ying Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China. .,The Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China.
| |
Collapse
|
23
|
Li D, Zheng L, Zhao D, Xu Y, Wang Y. The Role of Immune Cells in Recurrent Spontaneous Abortion. Reprod Sci 2021; 28:3303-3315. [PMID: 34101149 PMCID: PMC8186021 DOI: 10.1007/s43032-021-00599-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Recurrent spontaneous abortion affects approximately 1–2% of women of childbearing, and describes a condition in which women suffer from three or more continuous spontaneous miscarriages. However, the origin of recurrent spontaneous abortion (RSA) remains unknown, preventing effective treatment and placing stress upon patients. It has been acknowledged that successful pregnancy necessitates balanced immune responses. Therefore, immunological aberrancy may be considered a root cause of poor pregnancy outcomes. Considerable published studies have investigated the relationship between various immune cells and RSA. Here, we review current knowledge on this area, and discuss the five main categories of immune cells involved in RSA; these include innate lymphocytes (ILC), macrophages, decidual dendritic cells (DCs), and T cells. Furthermore, we sought to summarize the impact of the multiple interactions of various immune cells on the emergence of RSA. A good understanding of pregnancy-induced immunological alterations could reveal new therapeutic strategies for favorable pregnancy outcomes.
Collapse
Affiliation(s)
- Dan Li
- Reproductive Medical Center, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Reproductive Medical Center, The Second Hospital of Jilin University, Changchun, China
| | | | - Ying Xu
- Reproductive Medical Center, The Second Hospital of Jilin University, Changchun, China
| | - Yeling Wang
- Departments of Cardiovascular Medicine, First Hospital, Jilin University, Changchun, 130000, China.
| |
Collapse
|
24
|
Liu Y, Gao S, Zhao Y, Wang H, Pan Q, Shao Q. Decidual Natural Killer Cells: A Good Nanny at the Maternal-Fetal Interface During Early Pregnancy. Front Immunol 2021; 12:663660. [PMID: 34054831 PMCID: PMC8149889 DOI: 10.3389/fimmu.2021.663660] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
Decidual natural killer (dNK) cells are the tissue-resident and major subpopulation of NK cells at the maternal-fetal interface. It has been demonstrated that dNK cells play pivotal roles in pregnancy, including keeping maternal-fetal immune tolerance, promoting extravillous trophoblast (EVT) cell invasion, and driving uterine spiral artery remodeling. However, the molecular mechanisms haven't been elucidated until recent years. In this review, we systemically introduce the generation, subsets, and surface or soluble molecules of dNK cells, which are critical for maintaining the functions of dNK cells. Further, new functions of dNK cells including well-controlled cytotoxicity, immunosurveillance and immunotrophism supporting via the cell-cell interaction between dNK cells and EVT cells are mainly focused. The molecular mechanisms involved in these functions are also illustrated. Moreover, pregnancy-associated diseases caused by the dNK cells abnormalities are discussed. It will be important for future investigations about the mechanism of maintenance of pregnancy and parturition and potential clinical applications of dNK cells.
Collapse
Affiliation(s)
- Yuefang Liu
- Department of Clinical Genetics, the Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, China
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Zhenjiang, China
| | - Shujun Gao
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Zhenjiang, China
| | - Yangjing Zhao
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Zhenjiang, China
| | - Hui Wang
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Zhenjiang, China
| | - Qiong Pan
- Department of Clinical Genetics, the Huai'an Maternity and Child Clinical College of Xuzhou Medical University, Huai'an, China
| | - Qixiang Shao
- Reproductive Sciences Institute, Jiangsu University, Zhenjiang, China
- Department of Immunology, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Zhenjiang, China
- Jiangsu College of Nursing, School of Medical Science and Laboratory Medicine, Huai'an, China
| |
Collapse
|
25
|
Schust DJ, Bonney EA, Sugimoto J, Ezashi T, Roberts RM, Choi S, Zhou J. The Immunology of Syncytialized Trophoblast. Int J Mol Sci 2021; 22:ijms22041767. [PMID: 33578919 PMCID: PMC7916661 DOI: 10.3390/ijms22041767] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/02/2021] [Accepted: 02/06/2021] [Indexed: 02/06/2023] Open
Abstract
Multinucleate syncytialized trophoblast is found in three forms in the human placenta. In the earliest stages of pregnancy, it is seen at the invasive leading edge of the implanting embryo and has been called primitive trophoblast. In later pregnancy, it is represented by the immense, multinucleated layer covering the surface of placental villi and by the trophoblast giant cells found deep within the uterine decidua and myometrium. These syncytia interact with local and/or systemic maternal immune effector cells in a fine balance that allows for invasion and persistence of allogeneic cells in a mother who must retain immunocompetence for 40 weeks of pregnancy. Maternal immune interactions with syncytialized trophoblast require tightly regulated mechanisms that may differ depending on the location of fetal cells and their invasiveness, the nature of the surrounding immune effector cells and the gestational age of the pregnancy. Some specifically reflect the unique mechanisms involved in trophoblast cell–cell fusion (aka syncytialization). Here we will review and summarize several of the mechanisms that support healthy maternal–fetal immune interactions specifically at syncytiotrophoblast interfaces.
Collapse
Affiliation(s)
- Danny J. Schust
- Department of Obstetrics, Gynecology, University of Missouri School of Medicine, Columbia, MO 65202, USA; (T.E.); (R.M.R.); (S.C.); (J.Z.)
- Correspondence:
| | - Elizabeth A. Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont College of Medicine, Burlington, VT 05405, USA;
| | - Jun Sugimoto
- Department of Obstetrics and Gynecology, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Toshi Ezashi
- Department of Obstetrics, Gynecology, University of Missouri School of Medicine, Columbia, MO 65202, USA; (T.E.); (R.M.R.); (S.C.); (J.Z.)
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - R. Michael Roberts
- Department of Obstetrics, Gynecology, University of Missouri School of Medicine, Columbia, MO 65202, USA; (T.E.); (R.M.R.); (S.C.); (J.Z.)
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Sehee Choi
- Department of Obstetrics, Gynecology, University of Missouri School of Medicine, Columbia, MO 65202, USA; (T.E.); (R.M.R.); (S.C.); (J.Z.)
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Jie Zhou
- Department of Obstetrics, Gynecology, University of Missouri School of Medicine, Columbia, MO 65202, USA; (T.E.); (R.M.R.); (S.C.); (J.Z.)
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| |
Collapse
|
26
|
Recent insights into the impact of immune dysfunction on reproduction in autoimmune thyroiditis. Clin Immunol 2021; 224:108663. [PMID: 33401032 DOI: 10.1016/j.clim.2020.108663] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/22/2020] [Accepted: 12/30/2020] [Indexed: 02/06/2023]
Abstract
Autoimmune thyroiditis (AIT) is a common organ-specific autoimmune disease with a high incidence among women of childbearing age. Recent studies have reported that women with AIT are more susceptible to infertility, miscarriage and preterm birth. It has been investigated that abnormal changes in maternal immune system and maternal-fetal interface can dampen the immune tolerance between mother and fetus, which underlie the pathogenesis of adverse pregnancy outcomes. Hence, we summarize the immunological changes related to adverse reproductive outcomes in AIT and highlight the respective contributions of both humoral and cellular immune dysfunctions to pregnancy failures. Moreover, the direct impacts of AIT on maternal-fetal immune activation and biological influences to trophoblasts are discussed as well. All these associations require confirmation in larger studies, and the pathogenic mechanisms need to be better understood, which might provide useful information for clinical diagnosis and therapy of AIT.
Collapse
|
27
|
Wang Y, Li D, Yu T, Hu M, Xing J, Bai S, Qu W, Tong X. Dynamics of TIGIT and PD-1 expression on NK cells during the course of normal pregnancy. Immunol Lett 2020; 230:42-48. [PMID: 33359535 DOI: 10.1016/j.imlet.2020.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 01/16/2023]
Abstract
PROBLEM Immune checkpoint molecules are receptors that can transmit inhibitory signals into cells to negatively modulate the immune response. However, their roles in NK cells during normal pregnancy remain poorly understood. METHOD OF STUDY Peripheral blood samples were collected from women during the first, second and third trimesters of pregnancy. Peripheral blood NK (pNK) cells and T cells were analyzed for the expression of the immune checkpoint molecules TIGIT and PD-1 by flow cytometry. In addition, the ability of pNK cells to secret functional molecules was also evaluated. RESULTS The expression of TIGIT on pNK cells increased gradually throughout pregnancy, whereas that of PD-1 showed the opposite pattern. However, on T cells, the expression of both TIGIT and PD-1 peaked during early pregnancy, and then declined gradually thereafter. Moreover, the expressions of granzyme B, IFN-γ and CD107a by pNK cells also decreased over the course of pregnancy. Compared with TIGIT- NK cells, TIGIT + NK cells possessed reduced expression of functional molecules. CONCLUSIONS As pregnancy progressed, the levels of immune checkpoint molecules expressed on pNK cells and T cells changed and the two molecules showed different trends. Furthermore, the secretion of functional molecules from pNK cells was negatively correlated with the trend of TIGIT expression, indicating TIGIT may play an important role in modulating the functions of pNK cells during pregnancy. Further study of TIGIT expression on pNK cells may enhance our understanding of its role in maintaining maternal-fetal tolerance and provide a useful marker for predicting instability during pregnancy.
Collapse
Affiliation(s)
- Yanshi Wang
- Reproductive and Genetic Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Daojing Li
- Department of Clinical Laboratory, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Ting Yu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Meihong Hu
- Reproductive and Genetic Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Juan Xing
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shun Bai
- Reproductive and Genetic Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wanjun Qu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Xianhong Tong
- Reproductive and Genetic Hospital, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
28
|
Wang S, Li M, Sun F, Chen C, Ye J, Li D, Qian J, Du M. Th17/Treg-cell balance in the peripheral blood of pregnant females with a history of recurrent spontaneous abortion receiving progesterone or cyclosporine A. Exp Ther Med 2020; 21:37. [PMID: 33273967 DOI: 10.3892/etm.2020.9469] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 06/27/2019] [Indexed: 12/13/2022] Open
Abstract
A successful pregnancy requires the maternal immune system to accept a fetus expressing allogeneic paternal antigens and provide competent responses to infections. Accordingly, maternal-fetal immune abnormalities may have an important role in the development of recurrent spontaneous abortion (RSA). Ever since the establishment of the association between immunologic abnormalities and RSA, various types of immune therapy to restore normal immune homeostasis have been increasingly developed. Although previous studies have focused on the maternal-fetal interface, non-invasive examination is of great importance in clinical practice. The present study investigated the balance between type-17 T-helper (Th17) and T-regulatory (Treg) cells in the peripheral blood to improve the current understanding of the pathogenesis of RSA. Imbalances in Th17/Treg cells and associated molecular profiles were observed in patients with RSA. Furthermore, it was determined that the immunosuppressant cyclosporine A reduced the proportion of Th17 cells and promoted Treg-cell dominance by upregulating the expression of co-inhibitory molecules in pregnant females with a history of RSA. Progesterone, the traditional maternal-care drug, also had a certain immunomodulatory role through restoring the levels of several co-inhibitory molecules (including T-cell immunoglobulin mucin family member-3, programmed cell death-1 and cytotoxic T-lymphocyte associated protein-4) in the treatment of RSA. Changes in these immune molecules within the maternal peripheral blood may be indicators for monitoring pregnancy and prediction of RSA.
Collapse
Affiliation(s)
- Songcun Wang
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Shanghai 200080, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, P.R. China
| | - Mengdie Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Shanghai 200080, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, P.R. China
| | - Fengrun Sun
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Shanghai 200080, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, P.R. China
| | - Chunqin Chen
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Shanghai 200080, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, P.R. China
| | - Jiangfeng Ye
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, P.R. China
| | - Dajin Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Shanghai 200080, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, P.R. China
| | - Jinfeng Qian
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Shanghai 200080, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, P.R. China
| | - Meirong Du
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University Shanghai Medical College, Shanghai 200080, P.R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200080, P.R. China
| |
Collapse
|
29
|
Eikmans M, van der Zwan A, Claas FHJ, van der Hoorn ML, Heidt S. Got your mother in a whirl: The role of maternal T cells and myeloid cells in pregnancy. HLA 2020; 96:561-579. [PMID: 32841539 DOI: 10.1111/tan.14055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/09/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022]
Abstract
Appropriate development of the placenta is required for healthy pregnancy to occur. After implantation of the fertilized blastocyst, fetal trophoblasts invade the endometrium and myometrium of the mother's uterus to establish placentation. In this process, fetal trophoblasts encounter maternal immune cells. In this review, we focus on the role of maternal T cells and myeloid cells (macrophages, dendritic cells) in pregnancy and their interaction with trophoblasts. To retain immunologic tolerization, trophoblasts evade immune recognition by T cells and produce factors that modulate their phenotype and function. On top of that, the local environment at the maternal-fetal interface favors expansion of regulatory T cells. Macrophages and dendritic cells are essential in maintaining a healthy pregnancy. They produce soluble factors and act as antigen-presenting cells, thereby interacting with T cells. Herein, M2 macrophages, immature dendritic cells, CD4+ Th2 cells, and regulatory T cells represent an axis that maintains a local immune tolerant environment. We consider outstanding issues concerning these cell types and their pathways, which need to be addressed in future investigations. Data from recent single-cell sequencing experiments of the placental bed, to study heterogeneity of maternal immune cells and to predict cell-cell interactions, are discussed. Novel ways for long-term culturing of primary trophoblasts allow for cell-cell interaction studies in a functional way. Future directions should include study of the functionality of currently known and newly identified decidual immune cell subsets in healthy and complicated pregnancies, and their interaction with and modulation by trophoblast cells.
Collapse
Affiliation(s)
- Michael Eikmans
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anita van der Zwan
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frans H J Claas
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
30
|
Zinc Supplementation: Immune Balance of Pregnancy During the Chronic Phase of the Chagas Disease. Acta Parasitol 2020; 65:599-609. [PMID: 32141022 DOI: 10.2478/s11686-020-00188-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 02/18/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Chagas disease or American trypanosomiasis is caused by the protozoan Trypanosoma cruzi and is endemic of the Americas. The control of the disease is restricted to toxic and potentially teratogenic drugs, which limit the use during pregnancy. The use of food supplementation offers a safe and low-cost form to alleviate Chagas disease symptoms, mostly in areas with alimentary risk. For example, zinc demonstrates positive effects in immune response, including in Chagas disease during pregnancy. PURPOSE This study describes the innate response in pregnant rats chronically infected with T. cruzi and supplemented with zinc. METHODS Pregnant female Wistar rats, infected with T. cruzi, were treated with 20 mg/kg/day zinc sulfate and euthanized on the 18th day. Samples (plasma, splenocytes, and peritoneal exudate) were collected and several immune parameters (nitric oxide, RT1B, CD80/CD86, MCP-1, CD11b/c, NK/NKT, IL-2, IL-10, INF-cc, and apoptosis) evaluated. RESULTS Under Zinc supplementation and/or T. cruzi infection, the gestation developed normally. Several innate immune parameters such as RT1B, CD80/CD86, MCP-1 expressing lymphocytes, IL-2, and IL-17 were positively altered, whereas nitric oxide, CD11b/c, NK/NKT, apoptosis, INF-γ, and corticosterone demonstrated a pro-pregnancy pattern. CONCLUSION Our results indicated that zinc has diverse effects on immune response during pregnancy. An anti-T. cruzi immunity, as well as a pro-gestation response, were observed after zinc supplementation. The complete comprehension of zinc supplementation in pregnancy will base an adequate strategy to alleviate Chagas disease symptoms and propagation, especially for populations from endemic areas.
Collapse
|
31
|
Meggyes M, Nagy DU, Szereday L. Investigation of the PD-1 and PD-L1 Immune Checkpoint Molecules Throughout Healthy Human Pregnancy and in Nonpregnant Women. J Clin Med 2020; 9:jcm9082536. [PMID: 32781525 PMCID: PMC7464563 DOI: 10.3390/jcm9082536] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/30/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
Background: A growing body of evidence supports the importance of PD-1 and PD-L1, especially in the materno-fetal interface, although limited information is available about the peripheral expression of these molecules during the trimesters of pregnancy. Methods: 13 healthy women were enrolled from the 1st, 10 from the 2nd and 12 from the 3rd trimester of pregnancy at the same time, 10 healthy, age-matched nonpregnant women formed the control group. From peripheral blood, mononuclear cells were separated and stored at –80 °C. From freshly thawed samples, surface and intracellular staining were performed for flow cytometric analyses. CD107a degranulation assay was used to evaluate the cytotoxicity. Results: significant alternation was detected in PD-1 expression by CD8+T cells and in PD-L1 expression by CD8+T, CD4+T and Treg cells. An interesting relationship was revealed between the PD-1 and PD-L1 expression by the investigated subpopulations in 2nd trimester of pregnancy. Different expression patterns of an activation receptor NKG2D by the PD-1+ CD8+T cells was observed during pregnancy. The notable relationship was further determined in cytotoxicity between PD-1+ and NKG2D+ CD8+T cells throughout pregnancy. Conclusions: the different PD-1 presence and the relationship with NKG2D could contribute to the dynamic changes of the Th1 and Th2 predominance throughout the three trimesters of a healthy pregnancy.
Collapse
Affiliation(s)
- Matyas Meggyes
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 7624 Pecs, Hungary;
- Janos Szentagothai Research Centre, 7624 Pecs, Hungary
- Correspondence:
| | - David U. Nagy
- Medical Centre, Cochrane Hungary, University of Pécs, 7623 Pécs, Hungary;
| | - Laszlo Szereday
- Department of Medical Microbiology and Immunology, Medical School, University of Pecs, 7624 Pecs, Hungary;
- Janos Szentagothai Research Centre, 7624 Pecs, Hungary
| |
Collapse
|
32
|
Krop J, Heidt S, Claas FHJ, Eikmans M. Regulatory T Cells in Pregnancy: It Is Not All About FoxP3. Front Immunol 2020; 11:1182. [PMID: 32655556 PMCID: PMC7324675 DOI: 10.3389/fimmu.2020.01182] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
In pregnancy, the semi-allogeneic fetus needs to be tolerated by the mother's immune system. Regulatory T cells (Tregs) play a prominent role in this process. Novel technologies allow for in-depth phenotyping of previously unidentified immune cell subsets, which has resulted in the appreciation of a vast heterogeneity of Treg subsets. Similar to other immunological events, there appears to be great diversity within the Treg population during pregnancy, both at the maternal-fetal interface as in the peripheral blood. Different Treg subsets have distinct phenotypes and various ways of functioning. Furthermore, the frequency of individual Treg subsets varies throughout gestation and is altered in aberrant pregnancies. This suggests that distinct Treg subsets play a role at different time points of gestation and that their role in maintaining healthy pregnancy is crucial, as reflected for instance by their reduced frequency in women with recurrent pregnancy loss. Since pregnancy is essential for the existence of mankind, multiple immune regulatory mechanisms and cell types are likely at play to assure successful pregnancy. Therefore, it is important to understand the complete microenvironment of the decidua, preferably in the context of the whole immune cell repertoire of the pregnant woman. So far, most studies have focused on a single mechanism or cell type, which often is the FoxP3 positive regulatory T cell when studying immune regulation. In this review, we instead focus on the contribution of FoxP3 negative Treg subsets to the decidual microenvironment and their possible role in pregnancy complications. Their phenotype, function, and effect in pregnancy are discussed.
Collapse
Affiliation(s)
- Juliette Krop
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Sebastiaan Heidt
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Frans H J Claas
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Michael Eikmans
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
33
|
Zhao Y, Zheng Q, Jin L. The Role of B7 Family Molecules in Maternal-Fetal Immunity. Front Immunol 2020; 11:458. [PMID: 32265918 PMCID: PMC7105612 DOI: 10.3389/fimmu.2020.00458] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 02/27/2020] [Indexed: 01/08/2023] Open
Abstract
Pregnancy is a complex but well-arranged process, and a healthy fetus requires immune privilege and surveillance in the presence of paternally derived antigens. Maternal and fetal cells interact at the maternal–fetal interface. The upregulation and downregulation of maternal immunity executed by the leukocyte population predominantly depend on the activity of decidual natural killer cells and trophoblasts and are further modulated by a series of duplex signals. The B7 family, which consists of B7-1, B7-2, B7-H1, B7-DC, B7-H2, B7-H3, B7-H4, B7-H5, BTNL2, B7-H6, and B7-H7, is one of the most characterized and widely distributed signaling molecule superfamilies and conducts both stimulatory and inhibitory signals through separate interactions. In particular, the roles of B7-1, B7-2, B7-H1, and their corresponding receptors in the progression of normal pregnancy and some pregnancy complications have been extensively studied. Together with the TCR–MHC complex, B7 and its receptors play a critical role in cell proliferation and cytokine secretion. Depending on this ligand–receptor crosstalk, the balance between the tolerance and rejection of the fetus is perfectly maintained. This review aims to provide an overview of the current knowledge of the B7 family and its functions in regulating maternal–fetal immunity through individual interactions.
Collapse
Affiliation(s)
- Yongbo Zhao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qingliang Zheng
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liping Jin
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Ali S, Majid S, Niamat Ali M, Taing S. Evaluation of T cell cytokines and their role in recurrent miscarriage. Int Immunopharmacol 2020; 82:106347. [PMID: 32143004 DOI: 10.1016/j.intimp.2020.106347] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 02/08/2020] [Accepted: 02/23/2020] [Indexed: 01/03/2023]
Abstract
Recurrent miscarriage (RM) is defined as two or more consecutive pregnancy losses that affect approximately 5% of conceived women worldwide. RM is a multi-factorial reproductive problem and has been associated with parental chromosomal abnormalities, embryonic chromosomal rearrangements, uterine anomalies, autoimmune disorders, endocrine dysfunction, thrombophilia, life style factors, and maternal infections. However, the exact cause is still undecided in remaining 50% of cases. Immunological rejection of the embryo due to exacerbated maternal immune reaction against paternal embryonic antigens has been set forth as one of the significant reason for RM. The accurate means that shield the embryo during normal pregnancy from the attack of maternal immune network and dismissal are inadequately implicit. However, it is suggested that the genetically irreconcilable embryo escapes maternal immune rejection due to communication among many vital cytokines exuded at maternal-embryonic interface both by maternal and embryonic cells. Previous investigations suggested the Th1/Th2 dominance in altered immunity of RM patients, according to which the allogenic embryo flees maternal T cell reaction by inclining the Th0 differentiation toward Th2 pathway resulting into diminished pro-inflammatory Th1 immunity. However, recently pro-inflammatory Th17 cells and immunoregulatory Treg cells have been discovered as essential immune players in RM besides Th1/Th2 components. Cytokines are believed to develop a complicated regulatory network so as to establish a state of homeostasis between the semi-allogenic embryo and the maternal immune system. However, an adverse imbalance among cytokines at maternal-embryonic interface perhaps due to their gene polymorphisms may render immunoregulatory means not enough to re-establish homeostasis and thus may collapse pregnancy.
Collapse
Affiliation(s)
- Shafat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, 190006 Srinagar, J&K, India
| | - Sabhiya Majid
- Department of Biochemistry, Government Medical College, Srinagar, J&K, India
| | - Md Niamat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, 190006 Srinagar, J&K, India.
| | - Shahnaz Taing
- Department of Obstetrics and Gynaecology, Government Medical College Associated Lalla Ded Hospital, Srinagar, J&K, India
| |
Collapse
|
35
|
Zhang YH, Sun HX. Immune checkpoint molecules in pregnancy: Focus on regulatory T cells. Eur J Immunol 2020; 50:160-169. [PMID: 31953958 DOI: 10.1002/eji.201948382] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/15/2019] [Accepted: 01/16/2020] [Indexed: 12/22/2022]
Abstract
Regulatory T (Treg) cells are a specialized subpopulation of T cells that plays critical roles in the maintenance of immune homeostasis. Although efforts have been done, their role in human pregnancy is not fully understood. Numerous studies reported the presence of Treg cells throughout gestation by promoting maternal-fetal tolerance and fetal development. Furthermore, Treg population is heterogeneous as it is expressing different immune checkpoint molecules favoring immune suppressive function. Therefore, better understanding of the heterogeneity and function of Treg cells during pregnancy is critical for an effective immune intervention. Latest evidence has shown that several immune checkpoint molecules are closely associated with pregnancy outcome via multiple inhibitory mechanisms. Majority of these studies demonstrated the modulatory effects of immune checkpoint molecules on effector T-cell immunity, but their effects on Treg activation and function are still an enigma. In this review, we emphasize the potential influence of multiple immune checkpoint molecules, including CTLA-4, PD-1, Tim-3, LAG-3, and TIGIT, either in membrane or soluble form, on the function of decidual and peripheral Treg cells during pregnancy. Additionally, we discuss the promising future of targeting Treg cells via immune checkpoint molecules for pregnancy maintenance and prevention of complicated pregnancies.
Collapse
Affiliation(s)
- Yong-Hong Zhang
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, China
| | - Hai-Xiang Sun
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
36
|
Yeh CC, Yang MJ, Lussier EC, Tsai HW, Lo PF, Hsieh SL, Wang PH. Low plasma levels of decoy receptor 3 (DcR3) in the third trimester of pregnancy with preeclampsia. Taiwan J Obstet Gynecol 2019; 58:349-353. [PMID: 31122523 DOI: 10.1016/j.tjog.2019.03.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/12/2018] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVE The pathophysiology of preeclampsia, a major threat during pregnancy characterized by excessive inflammatory status, remains unclear. Decoy receptor 3 (DcR3), a soluble member of the tumor necrosis factor receptor (TNFR) superfamily, is capable of inducing anti-apoptosis via binding with TL1A and anti-inflammation by driving Th2 immune reactions. DcR3 may, therefore, play a role in immune modulation during pregnancy. The purpose of this study is to explore the role of DcR3 in normal and preeclamptic pregnancies. MATERIALS AND METHODS Plasma samples from 104 normal pregnant women (26, 42, and 36 in the first, second, and third trimester, respectively) and 10 patients with preeclampsia in the third trimester were collected. Plasma DcR3 levels were determined by using commercial ELISA kits. ANOVA and linear regression analysis were performed to analyze the relationship between gestational age and DcR3 levels. After adjusting for gestational days, the levels of plasma DcR3 in preeclamptic and non-preeclamptic women in the third trimester were compared. RESULTS The plasma levels of DcR3 gradually decreased as the gestational days increased during pregnancy (p < 0.05). In the third trimester, pregnant women with preeclampsia had significantly lower plasma DcR3 levels compared to non-preeclamptic women (p < 0.05). CONCLUSIONS We found that plasma DcR3 levels gradually decreased as gestation progressed. The levels of plasma DcR3 in preeclamptic women were significantly lower than those of normal pregnant women, suggesting that a potential involvement of DcR3 in normal pregnancy and decreased levels of DcR3 may be related to preeclampsia.
Collapse
Affiliation(s)
- Chang-Ching Yeh
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Jie Yang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Hsiao-Wen Tsai
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan; School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Pei-Fen Lo
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Nursing, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shie-Liang Hsieh
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Peng-Hui Wang
- Department of Obstetrics and Gynecology, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
37
|
Wang J, Liu C, Que W, Fujino M, Tong G, Yan H, Li XK. Immunomodulatory effects of Salvianolic acid B in a spontaneous abortion mouse model. J Reprod Immunol 2019; 137:103075. [PMID: 31918160 DOI: 10.1016/j.jri.2019.103075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 11/20/2019] [Accepted: 12/19/2019] [Indexed: 11/30/2022]
Abstract
Pregnancy is a kind of natural immune tolerance. Immune factors play an important role in recurrent spontaneous abortion and repeated implantation failure. Salvianolic acid B (SalB) has anti-tumor, anti-inflammatory, anti-oxidation and immunomodulatory functions. However, there are few reports on the relationship between SalB and maternal-fetal immune tolerance. In this study, CBA/J × DBA/2 J mice as a spontaneous abortion mouse model were given SalB. The results showed that the abortion rate was significantly decreased after SalB treatment. The populations of Nkp46 and cytotoxic CD8+ T cells in the placenta of female mice treated with SalB were significantly decreased. The qRT-PCR showed that SalB was able to significantly reduce the expression of pro-inflammatory factors and Toll-like Receptor in the placenta. In addition, SalB was able to increase the area of the labyrinth in the placenta. In conclusion, these findings suggest that SalB is beneficial for the immune-modulation at the maternal-fetal interface in a spontaneous abortion mouse model, resulting in a decrease in the abortion rate. This may encourage new ideas for the treatment of patients with repeated implantation failure.
Collapse
Affiliation(s)
- Jing Wang
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Chi Liu
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; Department of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weitao Que
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan; AIDS Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Guoqing Tong
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hua Yan
- Reproductive Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xiao-Kang Li
- Division of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| |
Collapse
|
38
|
Abstract
T cell immunoglobulin and mucin domain-containing protein 3 (TIM3), a member of the TIM family, was originally identified as a receptor expressed on interferon-γ-producing CD4+ and CD8+ T cells. Initial data indicated that TIM3 functioned as a 'co-inhibitory' or 'checkpoint' receptor, but due to the lack of a definable inhibitory signalling motif, it was also suggested that TIM3 might act as a co-stimulatory receptor. Recent studies have shown that TIM3 is part of a module that contains multiple co-inhibitory receptors (checkpoint receptors), which are co-expressed and co-regulated on dysfunctional or 'exhausted' T cells in chronic viral infections and cancer. Furthermore, co-blockade of TIM3 and programmed cell death 1 (PD1) can result in tumour regression in preclinical models and can improve anticancer T cell responses in patients with advanced cancers. Here, we highlight the developments in understanding TIM3 biology, including novel ligand identification and the discovery of loss-of-function mutations associated with human disease. In addition, we summarize emerging data from human clinical trials showing that TIM3 indeed acts as a 'checkpoint' receptor and that inhibition of TIM3 enhances the antitumour effect of PD1 blockade.
Collapse
|
39
|
Chen M, Gilbert N, Liu H. Reduced expression of PD-L1 in autoimmune thyroiditis attenuate trophoblast invasion through ERK/MMP pathway. Reprod Biol Endocrinol 2019; 17:86. [PMID: 31656199 PMCID: PMC6816196 DOI: 10.1186/s12958-019-0536-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/16/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Autoimmune thyroiditis (AIT) with euthyroid is associated with miscarriage. But the exact mechanism remains unclear. Studies have shown that the programmed cell death-1 (PD-1)/programmed cell death -ligand 1 (PD-L1) pathway is essential for normal pregnancy. However, the expression of PD-L1 in gestational trophoblasts in mice with autoimmune thyroiditis and the mechanisms leading to miscarriage have not been fully investigated. METHODS Immunofluorescence and Western blot were used to detect the expression of PD-L1, p-ERK, MMP-2 and MMP-9 in embryonic trophoblast cells of pregnant mice with AIT. The expression of PD-L1 in HTR-8/SVneo cells were silenced, and the expression of PD-L1, MMP-2, MMP-9, ERK and p-ERK1/2 was detected by Western blot analyses and immunofluorescence assays. Invasive assays were performed in PD-L1 silenced HTR-8/SVneo cells using a Transwell chamber. RESULTS Compared with normal pregnancy, the expression of PD-L1, ERK, p-ERK, MMP-2 and MMP-9 in embryonic trophoblast cells was significantly lower in pregnant mice with AIT. Compared with the negative control (NC) group (cells transfected with negative control siRNA), phosphorylation of MMP-2, MMP-9 and P-ERK1/2 proteins was significantly reduced in HTR-8/SVneo cells transfected with PD-L1 siRNA, and the number of cells penetrating the membrane was reduced. CONCLUSION AIT inhibits ERK/MMP-2 and MMP-9 pathways through PD-L1 reduction, attenuates embryonic trophoblast invasion and ultimalely induces miscarriage ultimately.
Collapse
Affiliation(s)
- Mengya Chen
- grid.452828.1Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027 Liaoning China
| | - Nduwimana Gilbert
- grid.452828.1Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027 Liaoning China
| | - Haixia Liu
- grid.452828.1Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116027 Liaoning China
| |
Collapse
|
40
|
Gurbuz RH, Atilla P, Orgul G, Tanacan A, Dolgun A, Cakar AN, Beksac MS. Impaired Placentation and Early Pregnancy Loss in Patients with MTHFR Polymorphisms and Type-1 Diabetes Mellitus. Fetal Pediatr Pathol 2019; 38:376-386. [PMID: 30955395 DOI: 10.1080/15513815.2019.1600623] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Objective: To evaluate the impact of type-1 diabetes mellitus (DM) and methylenetetrahydrofolate reductase (MTHFR) polymorphisms on impaired placentation leading to early pregnancy loss. Methods: Miscarriage materials were obtained from eight pregnant women with type-1 DM without MTHFR polymorphism, eight with MTHFR polymorphisms without type-1 DM, and eight controls with neither DM nor MTHFR polymorphisms. Insulin-like growth factor-1 (IGF-1), leukemia inhibitory factor (LIF), and Beclin-1 expression were assessed to evaluate placentation. Results: Cytoplasmic LIF, IGF-1, and Beclin-1 expression were decreased in the superficial and glandular epithelial cells of the decidua in both study groups. LIF expression was increased in interstitial trophoblasts in the MTHFR group. IGF-1 expression was decreased in the decidual cells and interstitial trophoblasts in both study groups, while the decrease in stromal cells was noted only in type-1 DM group. Beclin-1 expression was increased in interstitial and villous trophoblasts in both study groups. Conclusion: The expression of IGF-1, LIF, and Beclin-1 are altered in both the decidua and the trophoblasts in pregnancies of women with type-1 DM and MTHFR polymorphisms, compared to normal pregnancies undergoing (elective) terminations.
Collapse
Affiliation(s)
- Rumeysa Hekimoglu Gurbuz
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine , Ankara , Turkey
| | - Pergin Atilla
- Department of Histology and Embryology, Hacettepe University Faculty of Medicine , Ankara , Turkey
| | - Gokcen Orgul
- Department of Obstetrics and Gynecology, Division of Perinatology, Hacettepe University Faculty of Medicine , Ankara , Turkey
| | - Atakan Tanacan
- Department of Obstetrics and Gynecology, Division of Perinatology, Hacettepe University Faculty of Medicine , Ankara , Turkey
| | - Anil Dolgun
- College of Science, Engineering and Health, Lecturer of Statistics, RMIT University , Melbourne , Australia
| | - Ayse Nur Cakar
- Department of Histogy and Embryology, TOBB University Faculty of Medicine , Ankara , Turkey
| | - Mehmet Sinan Beksac
- Department of Obstetrics and Gynecology, Division of Perinatology, Hacettepe University Faculty of Medicine , Ankara , Turkey
| |
Collapse
|
41
|
Zhang Y, Ma L, Hu X, Ji J, Mor G, Liao A. The role of the PD-1/PD-L1 axis in macrophage differentiation and function during pregnancy. Hum Reprod 2019; 34:25-36. [PMID: 30500923 DOI: 10.1093/humrep/dey347] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 11/01/2018] [Indexed: 12/21/2022] Open
Abstract
STUDY QUESTION What is the role of the programmed cell death-1 (PD-1)/PD-1 ligand-1 (PD-L1) axis in macrophage polarization during early pregnancy? SUMMARY ANSWER PD-1 signaling is a major regulator of macrophage differentiation and function, and it is critical for the success of a pregnancy. WHAT IS KNOWN ALREADY The predominance of decidual macrophages (DMs) with an M2 phenotype is an important contributor to maternal-fetal tolerance during early pregnancy. STUDY DESIGN, SIZE, DURATION Twenty-four women with recurrent miscarriage (RM) and 70 women undergoing elective termination of an early normal pregnancy (NP) were included. Twelve female CBA/J, four male DBA/2, and four male BALB/c mice were included and mating carried out. The 12 CBA/J pregnant mice were then categorized into three groups of four mice: healthy control group CBA/J×BALB/c, abortion-prone pregnant group CBA/J×DBA/2 and normal pregnancies CBA/J×BALB/c treated with anti-PD-1 monoclonal antibodies. PARTICIPANTS/MATERIALS, SETTING, METHODS The profile of DMs, and the expression of PD-1 and PD-L1 in DMs from women with NP and RM were measured by flow cytometry. PD-L1 expression in human villi was determined by quantitative RT-PCR (qRT-PCR) and western blot. An in vitro model consisting of peripheral CD14+ monocytes isolated from women with NP was used. The profile of differentiated macrophages and their phagocytotic activity were then measured by flow cytometry. The mRNA levels of genes potentially underlying macrophage polarization modulated by PD-1 signaling were determined by qRT-PCR. Twelve pregnant mice were included in our in vivo model and underwent different treatment. The embryo resorption rate, and macrophage profile as well as PD-1 expression in murine spleens and uterus were analyzed by flow cytometry. MAIN RESULTS AND THE ROLE OF CHANCE Compared with NP, women with RM had elevated percentages of M1 DMs (P < 0.01), and reduced frequencies of M2 DMs (P < 0.05), as well as decreased PD-1 protein expression (P < 0.05) in the DMs. In addition, decreased mRNA and protein levels of PD-L1 expression in placental villi were observed in women with RM (P < 0.001). Using in vitro experiments, compared to the control group, we found that PD-1 activation by recombinant human (rh) PD-L1 Fc (human PD-L1 fused to the Fc region of human IgG1) drove the differentiation of macrophages with immuno-modulatory characteristics (P < 0.01). However, PD-1 blockade promoted dominance of the M1 phenotype (P < 0.01). PD-1 polarized macrophages showed enhanced phagocytic activity (P < 0.01), which was decreased with PD-1 blockade (P < 0.001). Furthermore, PD-1 blockade promoted the expression of pro-inflammatory cytokines and interferon regulatory factor (IRF) 5 (P < 0.05), while IRF4 expression was inhibited (P < 0.05). In addition, PD-1 blockade promoted macrophage glycolysis (P < 0.01) and inhibited fatty acid oxidation (P < 0.05). The mRNA expression levels of both phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin and mitogen-activated protein kinase/extracellular signal-regulated kinase/extracellular signal-regulated kinase were upregulated (P < 0.05) with PD-1 blockade during DM metabolic reprogramming. Moreover, in vivo mice data showed that PD-1 blockade or deficiency was associated with decreased M2 percentages at the maternal-fetal interface (P < 0.05) and embryo loss (P < 0.05). LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Whether the changes in DM polarization seen in miscarriage tissues are a cause or consequence of the demise of the pregnancy still requires further investigation. In addition, conducting metabolite analysis is required to further measure bioenergetic profiles. WIDER IMPLICATIONS OF THE FINDINGS This is the first study on the role of the PD-1/PD-L1 axis in macrophage polarization during early pregnancy; such exploration enhances our understanding of the physiology of early pregnancy. Our study also indicates that targeting the PD-1 pathway may represent a novel therapeutic strategy to prevent pregnancy loss. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by the National Nature Science Foundation of China (No. 81671490) and Integrated Innovative Team for Major Human Diseases Program of Tongji Medical College, HUST (No. 5001519002). None of the authors has any conflict of interest to declare.
Collapse
Affiliation(s)
- Yonghong Zhang
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China.,Division of Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Lina Ma
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xiaohui Hu
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Jinlu Ji
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Gil Mor
- Division of Reproductive Sciences, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Aihua Liao
- Family Planning Research Institute, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| |
Collapse
|
42
|
Miko E, Meggyes M, Doba K, Barakonyi A, Szereday L. Immune Checkpoint Molecules in Reproductive Immunology. Front Immunol 2019; 10:846. [PMID: 31057559 PMCID: PMC6482223 DOI: 10.3389/fimmu.2019.00846] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/01/2019] [Indexed: 01/11/2023] Open
Abstract
Immune checkpoint molecules, like CTLA-4, TIM-3, PD-1, are negative regulators of immune responses to avoid immune injury. Checkpoint regulators are thought to actively participate in the immune defense of infections, prevention of autoimmunity, transplantation, and tumor immune evasion. Maternal-fetal immunotolerance represents a real immunological challenge for the immune system of the mother: beside acceptance of the semiallogeneic fetus, the maternal immune system has to be prepared for immune defense mostly against infections. In this particular situation, the role of immune checkpoint molecules could be of special interest. In this review, we describe current knowledge on the role of immune checkpoint molecules in reproductive immunology.
Collapse
Affiliation(s)
- Eva Miko
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Pécs, Hungary.,Janos Szentagothai Research Centre, Pécs, Hungary
| | - Matyas Meggyes
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Pécs, Hungary.,Janos Szentagothai Research Centre, Pécs, Hungary
| | - Katalin Doba
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Pécs, Hungary
| | - Aliz Barakonyi
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Pécs, Hungary.,Janos Szentagothai Research Centre, Pécs, Hungary
| | - Laszlo Szereday
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Pécs, Hungary.,Janos Szentagothai Research Centre, Pécs, Hungary
| |
Collapse
|
43
|
Bukiya AN. Physiology of the Endocannabinoid System During Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1162:13-37. [PMID: 31332732 DOI: 10.1007/978-3-030-21737-2_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The endocannabinoid (eCB) system comprises endogenously produced cannabinoids (CBs), enzymes of their production and degradation, and CB-sensing receptors and transporters. The eCB system plays a critical role in virtually all stages of animal development. Studies on eCB system components and their physiological role have gained increasing attention with the rising legalization and medical use of marijuana products. The latter represent exogenous interventions that target the eCB system. This chapter summarizes knowledge in the field of CB contribution to gametogenesis, fertilization, embryo implantation, fetal development, birth, and adolescence-equivalent periods of ontogenesis. The material is complemented by the overview of data from our laboratory documenting the functional presence of the eCB system within cerebral arteries of baboons at different stages of development.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
44
|
Abstract
Multiple mechanisms of tolerance operate in the immune cross-talk at the fetomaternal interface, contributing to successful pregnancy outcome. The cross-talk includes interaction between various cell subsets and between cytokines and molecules of the endocrine system. A depiction of how all these components interact with each other and contribute to tolerance of the fetus is not clearly understood. Dysregulation in one or more of these mechanisms leads to fetal loss. Few effective biomarkers are available that can safely predict fetal loss. This review discusses some potential biomarkers that can predict failure of tolerance at the fetomaternal interface.
Collapse
Affiliation(s)
- Sudipta Tripathi
- Transplantation Research Center, Harvard Medical School, LMRC #316, 221 Longwood Avenue, Boston, MA 02115, USA
| | - Indira Guleria
- HLA Tissue Typing Laboratory, Renal Transplant Program, Division of Renal Medicine, Transplantation Research Center, Harvard Medical School, Brigham and Women's Hospital, 75 Francis Street, PBB 161G, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Streptococcus agalactiae Induces Placental Macrophages To Release Extracellular Traps Loaded with Tissue Remodeling Enzymes via an Oxidative Burst-Dependent Mechanism. mBio 2018; 9:mBio.02084-18. [PMID: 30459195 PMCID: PMC6247082 DOI: 10.1128/mbio.02084-18] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Streptococcus agalactiae, also known as group B Streptococcus (GBS), is a common pathogen during pregnancy where infection can result in chorioamnionitis, preterm premature rupture of membranes (PPROM), preterm labor, stillbirth, and neonatal sepsis. Mechanisms by which GBS infection results in adverse pregnancy outcomes are still incompletely understood. This study evaluated interactions between GBS and placental macrophages. The data demonstrate that in response to infection, placental macrophages release extracellular traps capable of killing GBS. Additionally, this work establishes that proteins associated with extracellular trap fibers include several matrix metalloproteinases that have been associated with chorioamnionitis. In the context of pregnancy, placental macrophage responses to bacterial infection might have beneficial and adverse consequences, including protective effects against bacterial invasion, but they may also release important mediators of membrane breakdown that could contribute to membrane rupture or preterm labor. Streptococcus agalactiae, or group B Streptococcus (GBS), is a common perinatal pathogen. GBS colonization of the vaginal mucosa during pregnancy is a risk factor for invasive infection of the fetal membranes (chorioamnionitis) and its consequences such as membrane rupture, preterm labor, stillbirth, and neonatal sepsis. Placental macrophages, or Hofbauer cells, are fetally derived macrophages present within placental and fetal membrane tissues that perform vital functions for fetal and placental development, including supporting angiogenesis, tissue remodeling, and regulation of maternal-fetal tolerance. Although placental macrophages as tissue-resident innate phagocytes are likely to engage invasive bacteria such as GBS, there is limited information regarding how these cells respond to bacterial infection. Here, we demonstrate in vitro that placental macrophages release macrophage extracellular traps (METs) in response to bacterial infection. Placental macrophage METs contain proteins, including histones, myeloperoxidase, and neutrophil elastase similar to neutrophil extracellular traps, and are capable of killing GBS cells. MET release from these cells occurs by a process that depends on the production of reactive oxygen species. Placental macrophage METs also contain matrix metalloproteases that are released in response to GBS and could contribute to fetal membrane weakening during infection. MET structures were identified within human fetal membrane tissues infected ex vivo, suggesting that placental macrophages release METs in response to bacterial infection during chorioamnionitis.
Collapse
|
46
|
Robinson KA, Orent W, Madsen JC, Benichou G. Maintaining T cell tolerance of alloantigens: Lessons from animal studies. Am J Transplant 2018; 18:1843-1856. [PMID: 29939471 PMCID: PMC6352985 DOI: 10.1111/ajt.14984] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 01/25/2023]
Abstract
Achieving host immune tolerance of allogeneic transplants represents the ultimate challenge in clinical transplantation. It has become clear that different cells and mechanisms participate in acquisition versus maintenance of allograft tolerance. Indeed, manipulations which prevent tolerance induction often fail to abrogate tolerance once it has been established. Hence, elucidation of the immunological mechanisms underlying maintenance of T cell tolerance to alloantigens is essential for the development of novel interventions that preserve a robust and long lasting state of allograft tolerance that relies on T cell deletion in addition to intra-graft suppression of inflammatory immune responses. In this review, we discuss some essential elements of the mechanisms involved in the maintenance of naturally occurring or experimentally induced allograft tolerance, including the newly described role of antigen cross-dressing mediated by extracellular vesicles.
Collapse
Affiliation(s)
- Kortney A. Robinson
- Center for Transplant Sciences, Massachusetts General
Hospital and Harvard Medical School, Boston, MA
| | - William Orent
- Center for Transplant Sciences, Massachusetts General
Hospital and Harvard Medical School, Boston, MA
| | - Joren C. Madsen
- Center for Transplant Sciences, Massachusetts General
Hospital and Harvard Medical School, Boston, MA.,Division of Cardiac Surgery, Department of Surgery,
Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Gilles Benichou
- Center for Transplant Sciences, Massachusetts General
Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
47
|
Zhou D, Zhi FJ, Qi MZ, Bai FR, Zhang G, Li JM, Liu H, Chen HT, Lin PF, Tang KQ, Liu W, Jin YP, Wang AH. Brucella induces unfolded protein response and inflammatory response via GntR in alveolar macrophages. Oncotarget 2017; 9:5184-5196. [PMID: 29435171 PMCID: PMC5797042 DOI: 10.18632/oncotarget.23706] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/04/2017] [Indexed: 01/18/2023] Open
Abstract
Brucella is an intracellular bacterium that causes the zoonosis brucellosis worldwide. Alveolar macrophages (AM) constitute the main cell target of inhaled Brucella. Brucella thwarts immune surveillance and evokes endoplasmic reticulum (ER) stress to replicate in macrophages via virulence factors. The GntR regulators family was concentrated as an important virulence factor in controlling virulence and intracellular survival of Brucella. However, the detailed underlying mechanism for the host-pathogen interaction is poorly understood. In this study the BSS2_II0438 mutant (ΔGntR) was constructed. The type IV secretion system (T4SS) virulence factor genes (VirB2, VirB6, and VirB8) were down-expression in ΔGntR. ΔGntR could infect and proliferate to high titers in GAMs without a significant difference compared with the parental strain. ΔGntR infection increased the expression of ER stress marker genes GRP78, ATF6, and PERK in the early stages of its intracellular cycle but decreased the expression of these genes in the late stages. ΔGntR increased greatly the number of Brucella CFUs in the inactive ER stress state in GAMs. Meanwhile, ΔGntR infection increased the levels of IFN-γ, IL-1β, and TNF-α, indicating ΔGntR could induce the secretion of inflammatory but not anti-inflammatory cytokines IL-10. Taken together, our results clarified the role of the GntR in B. suis. S2 virulence expression and elucidated that GntR is potentially involved in the signaling pathway of the Brucella-induced UPR and inflammatory response in GAMs.
Collapse
Affiliation(s)
- Dong Zhou
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Fei-Jie Zhi
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Mao-Zhen Qi
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Fu-Rong Bai
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Guangdong Zhang
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jun-Mei Li
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Huan Liu
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Hua-Tao Chen
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Peng-Fei Lin
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ke-Qiong Tang
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wei Liu
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ya-Ping Jin
- Key Laboratory of Animal Biotechnology of The Ministry of Agriculture, Northwest A&F University, Yangling, China.,College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ai-Hua Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
48
|
Chattopadhyay PK. What to expect when you're expecting. Sci Immunol 2017; 2:2/15/eaao3543. [PMID: 28864495 DOI: 10.1126/sciimmunol.aao3543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/09/2017] [Indexed: 11/02/2022]
Abstract
Applying systems immunology to study human pregnancy.
Collapse
Affiliation(s)
- Pratip K Chattopadhyay
- Department of Pathology and Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA.
| |
Collapse
|