1
|
Yang Y, Qiu J, Xu Q, Fan Y, Wang H, Qian H, Wu Z, Zhang Y, Gao Y, Shi C, Lu C, Xia Y, Cheng W. The loss of dNK1/2 and EVT1 cells at the maternal-fetal interface is associated with recurrent miscarriage†. Biol Reprod 2025; 112:119-129. [PMID: 39303127 DOI: 10.1093/biolre/ioae136] [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: 06/16/2024] [Revised: 08/24/2024] [Accepted: 09/19/2024] [Indexed: 09/22/2024] Open
Abstract
Recurrent miscarriage is a chronic and heterogeneous pregnancy disorder lacking effective treatment. Alterations at the maternal-fetal interface are commonly observed in recurrent miscarriage, with the loss of certain cell subpopulations believed to be a key cause. Through single-cell sequencing of recurrent miscarriage patients and healthy donors, we aim to identify aberrancy of cellular features in recurrent miscarriage tissues, providing new insights into the research. Natural killer cells, the most abundant immune cells in the decidua, are traditionally classified into dNK1, dNK2, and dNK3. In this study, we identified a new subset, dNK1/2, absent in recurrent miscarriage tissues. This subset was named because it expresses biomarkers of both dNK1 and dNK2. With further analysis, we discovered that dNK1/2 cells play roles in immunoregulation and cytokine secretion. On the villous side of the interface, a notable decrease of extravillous trophoblast cells was identified in recurrent miscarriage tissues. We clustered extravillous trophoblasts into EVT1 (absent in recurrent miscarriage) and EVT2 (retained in recurrent miscarriage). Pseudotime analysis revealed distinct differentiation paths, identifying CCNB1, HMGB1, and NPM1 as EVT1 biomarkers. Additionally, we found that EVT1 is involved in the regulation of cell death, while EVT2 exhibited more angiogenic activity. Cell communication analysis revealed that interaction between EVT1 and dNK1/2 mediates chemotaxis and endothelial cell regulation, crucial for spiral artery remodeling. The loss of this interaction may impair decidualization, which is associated with recurrent miscarriage. In summary, we propose that the loss of dNK1/2 and EVT1 cells is a significant pathological feature of recurrent miscarriage. SUMMARY SENTENCE The communication between EVT1 and dNK1/2 mediated the chemotaxis of EVT1 and facilitated regulation of endothelial cell death, initiating spiral artery remodeling. The loss of this specific cellular interaction may result in impaired decidualization, leading to recurrent miscarriage.
Collapse
Affiliation(s)
- Yijun Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Gynecology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Jiangnan Qiu
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qiaoqiao Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yun Fan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Wang
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hong Qian
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhu Wu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuchen Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yingchun Gao
- Department of Gynecology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Can Shi
- Department of Gynecology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Chuncheng Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wenjun Cheng
- Department of Gynecology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Mack JA, Burkholder A, Akhtari FS, House JS, Sovio U, Smith GCS, Schmitt CP, Fargo DC, Hall JE, Motsinger-Reif AA. A multi-ancestry genome-wide association study identifies novel candidate loci in the RARB gene associated with hypertensive disorders of pregnancy. HGG ADVANCES 2025; 6:100385. [PMID: 39580622 PMCID: PMC11667702 DOI: 10.1016/j.xhgg.2024.100385] [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: 05/30/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Genetic factors related to pregnancy-related traits are understudied, especially in ancestrally diverse cohorts. To assess maternal contributions to hypertensive disorders of pregnancy (HDP), we performed a multi-ancestry genome-wide association study (GWAS) of HDP in data from the North Carolina-based Personalized Environment and Genes Study (PEGS) cohort with validation in the UK Biobank (UKBB). The GWAS revealed two maternal loci associated with HDP at the genome-wide significance level. The lead independent variants were rs114954125 on chromosome 2 (near LRP1B; odds ratio [OR] [95% confidence interval {CI}]): 2.96 [2.02-4.34]; p = 2.82 × 10-8) and rs61176331 on chromosome 3 (on RARB; OR (95% CI): 3.08 (2.12-4.48); p = 3.52 × 10-9). We validated the associations near RARB with a meta-analysis of PEGS and the UKBB. We also identified cis-expression quantitative trait loci in the candidate region associated with decreased RARB expression in macrophage cells exposed to Salmonella. Chromatin mapping in FUMA identified a significant interaction within chromosome 3's enhancer and open chromatin regions, with strong effects observed for RARB and H3P10 gene regulation in mesendoderm cells, mesenchymal stem cells, and trophoblast-like stem cells. We applied existing polygenic scores (PGS) for preeclampsia and gestational hypertension and found that the scores were significantly associated with HDP in PEGS. The findings demonstrate the power of multi-ancestry studies for genetic discovery and highlight the relationship between immune response, regulation, and HDP and the utility of PGS for risk prediction. (PEGS is registered at ClinicalTrials.gov: NCT00341237.).
Collapse
Affiliation(s)
- Jasmine A Mack
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA; University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Adam Burkholder
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Farida S Akhtari
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - John S House
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Ulla Sovio
- University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Gordon C S Smith
- University of Cambridge, Department of Obstetrics and Gynaecology, Cambridge CB2 0SW, UK
| | - Charles P Schmitt
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - David C Fargo
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Janet E Hall
- National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | | |
Collapse
|
3
|
Yue S, Meng J. Role of Decidual Natural Killer Cells in the Pathogenesis of Preeclampsia. Am J Reprod Immunol 2025; 93:e70033. [PMID: 39739937 DOI: 10.1111/aji.70033] [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: 10/11/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
Preeclampsia is one of the most severe obstetric complications, yet its pathogenesis remains unclear. Decidual natural killer (dNK) cells, the most abundant immune cells at the maternal-fetal interface, are closely associated with preeclampsia due to abnormalities in their quantity, phenotype, and function. This review summarizes the molecular mechanisms by which dNK cells regulate extravillous trophoblast (EVT) invasion, promote uterine spiral artery remodeling, and maintain immune tolerance. Furthermore, it explores how disruptions in these mechanisms and changes in the decidual microenvironment alter dNK cell properties, driving the progression of preeclampsia. Understanding the mechanisms of dNK cells and identifying potential therapeutic targets may provide new insights for clinical intervention.
Collapse
Affiliation(s)
- Shuang Yue
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jinlai Meng
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, China
| |
Collapse
|
4
|
Gan J, Yang L, Yang SH, Gu WW, Gu Y, Shi Y, Shi JX, Xu HR, Xin YW, Zhang X, Wang J. FXYD1 was identified as a hub gene in recurrent miscarriage and involved in decidualization via regulating Na/K-ATPase activity. J Assist Reprod Genet 2024:10.1007/s10815-024-03363-8. [PMID: 39730944 DOI: 10.1007/s10815-024-03363-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/13/2024] [Indexed: 12/29/2024] Open
Abstract
PURPOSE Recurrent miscarriage (RM) is a distressing and complicated adverse pregnancy outcome. It is commonly recognized that insufficient decidualization could result in RM, but the molecular mechanisms of decidual impairment are still not fully understood. Thus, this study aimed to identify novel key genes potentially involved in RM and explore their roles played in endometrial decidualization. METHODS Initially, a combinative analysis of decidual and mid-secretory endometrial transcriptomes was performed to discover hub genes involved in the etiology of RM. And the expression levels of hub genes were evaluated in both primary decidual stromal cells (DSCs) and decidual tissues. Subsequently, the immortalized human endometrial cell line, T-HESCs, was used to investigate whether FXYD1 overexpression affects decidualization by regulating Na/K-ATPase activity. RESULTS FXYD domain containing ion transport regulator 1 (FXYD1) was identified as a hub gene in the pathogenesis of RM through various bioinformatic methods. Abnormally increased FXYD1 expression was observed in DSCs and decidual tissues from RM patients compared to that of the normal group. Furthermore, in vitro decidualization was obviously inhibited by the overexpression of FXYD1. Additionally, Na/K-ATPase activity was significantly elevated during decidualization, whereas overexpression of FXYD1 reduced Na/K-ATPase activity. Bufalin, a Na/K-ATPase inhibitor, showed an effectively inhibitory effect on decidualization. CONCLUSIONS Collectively, FXYD1 was discovered as a hub gene associated with RM, and its expression levels in RM patients were significantly upregulated. Increased FXYD1 expression might lead to decidualization defects by reducing Na/K-ATPase activity, of which presented a novel prospective treatment target for RM.
Collapse
Affiliation(s)
- Jie Gan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China
| | - Long Yang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China
| | - Shu-Han Yang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China
| | - Wen-Wen Gu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China
| | - Yan Gu
- The Second Hospital of Tianjin Medical University, Tianjin, 300221, China
| | - Yan Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China
| | - Jia-Xin Shi
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China
| | - Hao-Ran Xu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China
| | - Ya-Wei Xin
- The Second Hospital of Tianjin Medical University, Tianjin, 300221, China
| | - Xuan Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China.
| | - Jian Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of ReproductionRegulation,Shanghai Institute for Biomedical and Pharmaceutical Technologies,Medical School, Fudan University, Shanghai, 200237, China.
| |
Collapse
|
5
|
Kindschuh WF, Austin GI, Meydan Y, Park H, Urban JA, Watters E, Pollak S, Saade GR, Chung J, Mercer BM, Grobman WA, Haas DM, Silver RM, Serrano M, Buck GA, McNeil R, Nandakumar R, Reddy U, Wapner RJ, Kav AB, Uhlemann AC, Korem T. Early prediction of preeclampsia using the first trimester vaginal microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.01.626267. [PMID: 39677801 PMCID: PMC11642775 DOI: 10.1101/2024.12.01.626267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Preeclampsia is a severe obstetrical syndrome which contributes to 10-15% of all maternal deaths. Although the mechanisms underlying systemic damage in preeclampsia-such as impaired placentation, endothelial dysfunction, and immune dysregulation-are well studied, the initial triggers of the condition remain largely unknown. Furthermore, although the pathogenesis of preeclampsia begins early in pregnancy, there are no early diagnostics for this life-threatening syndrome, which is typically diagnosed much later, after systemic damage has already manifested. Here, we performed deep metagenomic sequencing and multiplex immunoassays of vaginal samples collected during the first trimester from 124 pregnant individuals, including 62 who developed preeclampsia with severe features. We identified multiple significant associations between vaginal immune factors, microbes, clinical factors, and the early pathogenesis of preeclampsia. These associations vary with BMI, and stratification revealed strong associations between preeclampsia and Bifidobacterium spp., Prevotella timonensis, and Sneathia vaginalis. Finally, we developed machine learning models that predict the development of preeclampsia using this first trimester data, collected ~5.7 months prior to clinical diagnosis, with an auROC of 0.78. We validated our models using data from an independent cohort (MOMS-PI), achieving an auROC of 0.80. Our findings highlight robust associations among the vaginal microbiome, local host immunity, and early pathogenic processes of preeclampsia, paving the way for early detection, prevention and intervention for this devastating condition.
Collapse
Affiliation(s)
- William F. Kindschuh
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - George I. Austin
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, NY, USA
| | - Yoli Meydan
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Heekuk Park
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Julia A. Urban
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Emily Watters
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Susan Pollak
- Biomarkers Core, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - George R. Saade
- Department of Obstetrics and Gynecology, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Judith Chung
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of California Irvine, CA, USA
| | - Brian M. Mercer
- Departments of Obstetrics and Gynecology, Case Western Reserve University, Cleveland, OH, USA
| | | | - David M. Haas
- Department of Obstetrics and Gynecology, Indiana University, Indianapolis, IN, USA
| | - Robert M. Silver
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA
| | - Myrna Serrano
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, VA, USA
| | - Gregory A. Buck
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Center for Microbiome Engineering and Data Analysis, Virginia Commonwealth University, Richmond, VA, USA
- Department of Computer Science, School of Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | | | - Renu Nandakumar
- Biomarkers Core, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Uma Reddy
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ronald J. Wapner
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Aya Brown Kav
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Anne-Catrin Uhlemann
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Division of Infectious Diseases, Columbia University Irving Medical Center, New York, NY, USA
| | - Tal Korem
- Program for Mathematical Genomics, Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
6
|
Brooker IA, Fisher JJ, Sutherland JM, Pringle KG. Understanding the impact of placental oxidative and nitrative stress in pregnancies complicated by fetal growth restriction. Placenta 2024; 158:318-328. [PMID: 39577026 DOI: 10.1016/j.placenta.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/24/2024]
Abstract
Fetal growth restriction (FGR) impacts approximately 10 % of all pregnancies worldwide and is associated with major adverse effects on fetal health in both the short- and long-term [1]. FGR most commonly arises as a result of impaired placentation, occurring in up to 60 % of cases in developed countries [2]. This narrative review outlines the impact of defective placentation on the placenta, focusing on redox imbalance, how this leads to placental oxidative and nitrative stress, and the implications of these stressors on placental nutrient transfer, premature replicative senescence, and trophoblast cell death. Furthermore, this review highlights the pivotal role of antioxidants in protecting against oxidative and nitrative damage by reducing the burden of reactive species. We explore how targeting antioxidants in pregnancy provides a promising strategy for preventing or treating FGR, to ultimately reduce the devastating burden of FGR on infant health.
Collapse
Affiliation(s)
- India A Brooker
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Joshua J Fisher
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Mothers and Babies Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jessie M Sutherland
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Kirsty G Pringle
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia; Women's Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.
| |
Collapse
|
7
|
Liu J, Dong Y, Zhou Y, Wang W, Li Y, Pei J. Exploring genetic associations between immune cells and hypertensive disorder of pregnancy using Mendelian randomization. BMC Pregnancy Childbirth 2024; 24:756. [PMID: 39548401 PMCID: PMC11566496 DOI: 10.1186/s12884-024-06950-w] [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: 05/16/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Observational epidemiological studies suggested that immunological dysregulation and inflammation play a significant role in the placental and renal dysfunction that leads to maternal hypertension. The immunophenotypes' possible causalities with hypertensive disease of pregnancy remain ambiguous. We performed two-sample Mendelian randomization (MR) analyses to comprehensively investigate the causal effect of immunophenotypes on hypertensive disorder of pregnancy (HDP). METHODS The large-scale genome-wide association studies (GWASs) data on immunological traits was taken from public catalog for 731 immunophenotypes. The summarized GWAS data in 4 types of HDP were retrieved from FinnGen database, including 811,605 Finnish individuals. The primary analysis was the inverse variance weighted (IVW) method, supplemented by conducting sensitivity analysis. To confirm whether cardiovascular proteins mediated the causal effect of immune cells on HDP, we additionally executed a mediation MR study. RESULTS After looking into genetically predicted immunophenotype biomarkers, we discovered 14 highly correlative immunophenotypes and 104 suggestive possible factors. The IVW analysis indicated that HLA DR on myeloid DC, HLA DR on plasmacytoid DC, and HLA DR on DC had a significant association with pre-eclampsia/eclampsia (PE), whereas CD4+ CD8dim AC and CD4+ CD8dim % leukocyte were protective against gestational hypertension (GH). All of HDP in our study had no statistically significant impact on immune cells, according to reverse MR analysis. The mediating role of LOX-1between HLA DR on plasmacytoid DC and chronic hypertension prior to pregnancy was validated. CONCLUSION This study showed that many immunophenotypes are implicated in HDP. Furthermore, the level of LOX-1 mediated the pathophysiology relationship between HLA DR on plasmacytoid dendritic cells and chronic hypertension prior to pregnancy.
Collapse
Affiliation(s)
- Jingting Liu
- Maternal and Child Health Care Research Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, 730050, China
| | - Yijun Dong
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730000, China
| | - Yawei Zhou
- Maternal and Child Health Care Research Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, 730050, China
| | - Wendi Wang
- Maternal and Child Health Care Research Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, 730050, China
| | - Yan Li
- Department of Biochemistry and Molecular Biology, Medical College of Northwest Minzu University, Lanzhou, 730030, China.
| | - Jianying Pei
- Maternal and Child Health Care Research Center, Gansu Provincial Maternity and Child Care Hospital, Lanzhou, 730050, China.
| |
Collapse
|
8
|
Asiimwe R, Knott B, Greene ME, Wright E, Bell M, Epstein D, Yates SD, Gonzalez MV, Fry S, Boydston E, Clevenger S, Locke JE, Brocato BE, Burgan CM, Burney R, Arora N, Duncan VE, Richter HE, Gunn D, Freud AG, Little SC, Porrett PM. Inhibition of NFAT promotes loss of tissue resident uterine natural killer cells and attendant pregnancy complications in humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583906. [PMID: 38559147 PMCID: PMC10979847 DOI: 10.1101/2024.03.07.583906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Uterine natural killer cells (uNKs) are a tissue resident lymphocyte population that are critical for pregnancy success. Although mouse models have demonstrated that NK deficiency results in abnormal placentation and poor pregnancy outcomes, the generalizability of this knowledge to humans remains unclear. Here we identify uterus transplant (UTx) recipients as a human population with reduced uNK cells and altered pregnancy phenotypes. We show that the NK reduction in UTx correlates with impaired transcriptional programming of NK tissue residency arising from the inhibition of NFAT-mediated signaling. Our observations suggest that NFAT-dependent genes modulate multiple molecular tissue residency programs in uNKs. These include early residency programs involving AP-1-family transcription factors and TGF-β-mediated upregulation of surface integrins. Collectively, our data identify a previously undescribed role for NFAT in uterine NK tissue residency and provide novel mechanistic insights into the biologic basis of pregnancy complications due to alteration of tissue resident NK subsets in humans. One Sentence Summary Role of NFAT in uterine NK cell tissue residency.
Collapse
|
9
|
Brouwers L, de Gier S, Vogelvang TE, Veerbeek JHW, Franx A, van Rijn BB, Nikkels PGJ. Prevalence of placental bed spiral artery pathology in preeclampsia and fetal growth restriction: A prospective cohort study. Placenta 2024; 156:1-9. [PMID: 39214009 DOI: 10.1016/j.placenta.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Preeclampsia and fetal growth restriction (PE/FGR) are pregnancy complications known to be associated with poor utero-placental function due to abnormal "physiological" remodeling of spiral arteries and unfavorable maternal cardiovascular health. However, the prevalence and degree of impaired spiral artery remodeling has not been clearly established. METHOD Prospective, multi-center observational cohort study to assess the prevalence of lesions associated with abnormal development of spiral arteries in placental bed biopsies systematically obtained from 121 women undergoing Caesarian section for PE/FGR compared with a reference group of 149 healthy controls. RESULTS PE/FGR was associated with a high prevalence of impaired spiral artery remodeling compared with controls (63.6 vs 10.1 %, p < 0.001), and a higher prevalence of non-remodeled spiral arteries without the presence of intramural trophoblast (45.5 vs 6.7 %, p < 0.001), despite abundant interstitial trophoblast invasion in surrounding decidua and myometrium. Normal remodeling was associated with circumferential presence of intramural trophoblast and hardly any trophoblast in surrounding tissue. Acute atherosis (28.9 vs 3.4 %, p < 0.001) and thrombosis (16.5 vs 5.4 %, p = 0.003) lesions were significantly more prevalent in PE/FGR. Impaired remodeling, acute atherosis and thrombosis lesions were equally present in both decidual and myometrial segments of the spiral arteries in both groups. Impaired remodeling was most prominent in the groups with FGR (with or without PE) and thrombosis was most often seen in the group with PE and FGR. CONCLUSION PE/FGR is associated with a high prevalence of impaired physiological remodeling and vascular lesions of the uterine spiral arteries in the placental bed.
Collapse
Affiliation(s)
- Laura Brouwers
- Wilhelmina Children's Hospital Birth Center, Department of Obstetrics, Division Woman and Baby, University Medical Center Utrecht, PO Box 85090, 3508 AB, Utrecht, the Netherlands
| | - Steffie de Gier
- Department of Pathology, University Medical Center Utrecht, Utrecht University, PO Box 85090, 3508 AB, Utrecht, the Netherlands
| | - Tatjana E Vogelvang
- Department of Gynecology & Obstetrics, Diakonessenhuis Hospital, PO Box 80250, 3508 TG, Utrecht, the Netherlands
| | - Jan H W Veerbeek
- Department of Gynecology & Obstetrics, Diakonessenhuis Hospital, PO Box 80250, 3508 TG, Utrecht, the Netherlands
| | - Arie Franx
- Wilhelmina Children's Hospital Birth Center, Department of Obstetrics, Division Woman and Baby, University Medical Center Utrecht, PO Box 85090, 3508 AB, Utrecht, the Netherlands; Division of Obstetrics & Gynecology, Department of Obstetrics and Fetal Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - Bas B van Rijn
- Wilhelmina Children's Hospital Birth Center, Department of Obstetrics, Division Woman and Baby, University Medical Center Utrecht, PO Box 85090, 3508 AB, Utrecht, the Netherlands; Department of Obstetrics and Gynecology, Maxima Medical Center, PO Box 7777, 5500 MB, Veldhoven, the Netherlands; Department of Biomedical Engineering, Eindhoven Technical University, PO Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Peter G J Nikkels
- Department of Pathology, University Medical Center Utrecht, Utrecht University, PO Box 85090, 3508 AB, Utrecht, the Netherlands.
| |
Collapse
|
10
|
Anima B, Gurusubramanian G, Roy VK. Hormonal dependent expression of apelin and apelin receptor in the ovary and uterus of mice. Reprod Biol 2024; 24:100918. [PMID: 38924877 DOI: 10.1016/j.repbio.2024.100918] [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: 03/05/2024] [Revised: 05/24/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Apelin and APJ have been shown to regulate female reproductive functions. However, its uterine expression during the oestrous cycle and its regulation by ovarian steroids, along with gonadotropin regulation in the ovary, has not been investigated. This study aimed to analyze the steroid-dependent uterine expression of apelin/APJ in the uterus along with the oestrous cycle. Furthermore, it also aimed to investigate gonadotropin-dependent ovarian expression of apelin and APJ. To investigate the uterine expression of apelin and APJ during estrous cycle in mice, uterus at different estrous stage were collected. To explore the ovarian steroids dependent expression of apelin system in the uterus, ovariectomized mice were treated with only estrogen at dose of 30 ng/g, only progesterone at dose of 150 μg/g and combined doses. To study the effect of gonadotropin on ovarian expression of apelin system, immature mice were injected with 2.5 IU of pregnant mare serum gonadotropin (PMSG) alone and both PMSG plus 2.5 IU of chorionic gonadotropin (hCG). Apelin and APJ protein expression are modulated by estrous phases in the uterus. The uterine apelin and APJ expression are up-regulated by estrogen and down-regulated by progesterone. The expression and localization of APJ showed increased abundance in the follicles of PMSG treated mice, however, the PMSG plus HCG treatment showed formation of corpus luteum with increased abundance of APJ and progesterone secretion. The expression of apelin and APJ are regulated by pituitary gonadotropin in the ovary and uterine apelin system by ovarian steroid hormone.
Collapse
Affiliation(s)
- Borgohain Anima
- Department of Zoology, Mizoram University, Aizawl 796004, Mizoram, India
| | | | - Vikas Kumar Roy
- Department of Zoology, Mizoram University, Aizawl 796004, Mizoram, India.
| |
Collapse
|
11
|
Li MY, Shen HH, Cao XY, Gao XX, Xu FY, Ha SY, Sun JS, Liu SP, Xie F, Li MQ. Targeting a mTOR/autophagy axis: a double-edged sword of rapamycin in spontaneous miscarriage. Biomed Pharmacother 2024; 177:116976. [PMID: 38906022 DOI: 10.1016/j.biopha.2024.116976] [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: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Immune dysfunction is a primary culprit behind spontaneous miscarriage (SM). To address this, immunosuppressive agents have emerged as a novel class of tocolytic drugs, modulating the maternal immune system's tolerance towards the embryo. Rapamycin (PubChem CID:5284616), a dual-purpose compound, functions as an immunosuppressive agent and triggers autophagy by targeting the mTOR pathway. Its efficacy in treating SM has garnered significant research interest in recent times. Autophagy, the cellular process of self-degradation and recycling, plays a pivotal role in numerous health conditions. Research indicates that autophagy is integral to endometrial decidualization, trophoblast invasion, and the proper functioning of decidual immune cells during a healthy pregnancy. Yet, in cases of SM, there is a dysregulation of the mTOR/autophagy axis in decidual stromal cells or immune cells at the maternal-fetal interface. Both in vitro and in vivo studies have highlighted the potential benefits of low-dose rapamycin in managing SM. However, given mTOR's critical role in energy metabolism, inhibiting it could potentially harm the pregnancy. Moreover, while low-dose rapamycin has been deemed safe for treating recurrent implant failure, its potential teratogenic effects remain uncertain due to insufficient data. In summary, rapamycin represents a double-edged sword in the treatment of SM, balancing its impact on autophagy and immune regulation. Further investigation is warranted to fully understand its implications.
Collapse
Affiliation(s)
- Meng-Ying Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Hui-Hui Shen
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Yan Cao
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Xiao Gao
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, People's Republic of China
| | - Feng-Yuan Xu
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| | - Si-Yao Ha
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510235, China
| | - Jian-Song Sun
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, People's Republic of China
| | - Song-Ping Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, People's Republic of China.
| | - Feng Xie
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China.
| | - Ming-Qing Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Department of Gynecologic Endocrinology and Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, People's Republic of China.
| |
Collapse
|
12
|
Wang H, Wang L, Gong G, Lin X, Luo J, Liu C, Mor G, Liao A. Interleukin-10: a novel metabolic inducer of macrophage differentiation and subsequently contributing to improved pregnancy outcomes of mice by orchestrating oxidative phosphorylation metabolism†. Biol Reprod 2024; 111:76-91. [PMID: 38501817 PMCID: PMC11466864 DOI: 10.1093/biolre/ioae041] [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: 08/21/2023] [Revised: 12/14/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Metabolism regulates the phenotype and function of macrophages. After recruitment to local tissues, monocytes are influenced by the local microenvironment and differentiate into various macrophages depending on different metabolic pathways. However, the metabolic mechanisms underlying decidual macrophage differentiation remain unknown. Interleukin-10 (IL-10) is an important decidual macrophage inducer and promotes oxidative phosphorylation (OXPHOS) of bone marrow-derived macrophages. In this study, we mainly investigate the metabolic changes involved in IL-10-generated macrophages from monocytes using in vitro models. We demonstrate that exposure of monocytes (either peripheral or THP-1) to IL-10 altered the phenotype and function of resultant macrophages that are linked with OXPHOS changes. Interleukin-10 enhanced the mitochondrial complex I and III activity of THP-1 cell-differentiated macrophages and increased the mitochondrial membrane potential, intracellular adenosine triphosphate, and reactive oxygen species levels. Oxidative phosphorylation blockage with oligomycin changed the cell morphology of IL-10-generated macrophages and the expression levels of cytokines, such as transforming growth factor beta, tumor necrosis factor-alpha, interferon gamma, and IL-10, apart from changes in the expression level of the surface markers CD206, CD209, and CD163. Moreover, in vivo IL-10 administration reduced the lipopolysaccharide (LPS)-induced embryo resorption rate, and this effect was diminished when OXPHOS was inhibited, demonstrating that OXPHOS is important for the improved pregnancy outcomes of IL-10 in LPS-induced abortion-prone mice. Our findings provide deep insights into the roles of IL-10 in macrophage biology and pregnancy maintenance. Nevertheless, the direct evidence that OXPHOS is involved in decidual macrophage differentiation needs further investigations.
Collapse
Affiliation(s)
- Huan Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Liling Wang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Guangshun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Xinxiu Lin
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Jing Luo
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Chunyan Liu
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Gil Mor
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
- C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Aihua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| |
Collapse
|
13
|
Wang X, Wang S, Xu X, Jiang Y, Ren L, Zhang H, Li Z, Liu X, Hu X, Ren Y. The effect of Toxoplasma gondii infection on galectin-9 expression in decidual macrophages contributing to dysfunction of decidual NK cells during pregnancy. Parasit Vectors 2024; 17:299. [PMID: 38987795 PMCID: PMC11234737 DOI: 10.1186/s13071-024-06379-2] [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: 05/17/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Toxoplasma gondii infection causes adverse pregnancy outcomes by affecting the expression of immunotolerant molecules in decidual immune cells. Galectin-9 (Gal-9) is widely expressed in decidual macrophages (dMφ) and is crucial for maintaining normal pregnancy by interacting with the immunomodulatory protein T-cell immunoglobulin and mucin domain-containing molecule 3 (Tim-3). However, the effects of T. gondii infection on Gal-9 expression in dMφ, and the impact of altered Gal-9 expression levels on the maternal-fetal tolerance function of decidual natural killer (dNK) cells, are still unknown. METHODS Pregnancy outcomes of T. gondii-infected C57BL/6 and Lgals9-/- pregnant mice models were recorded. Expression of Gal-9, c-Jun N-terminal kinase (JNK), phosphorylated JNK (p-JNK), and Forkhead box protein O1 (FOXO1) was detected by western blotting, flow cytometry or immunofluorescence. The binding of FOXO1 to the promoter of Lgals9 was determined by chromatin immunoprecipitation-polymerase chain reaction (ChIP-PCR). The expression of extracellular signal-regulated kinase (ERK), phosphorylated ERK (p-ERK), cAMP-response element binding protein (CREB), phosphorylated CREB (p-CREB), T-box expressed in T cells (T-bet), interleukin 10 (IL-10), and interferon gamma (IFN-γ) in dNK cells was assayed by western blotting. RESULTS Toxoplasma gondii infection increased the expression of p-JNK and FOXO1 in dMφ, resulting in a reduction in Gal-9 due to the elevated binding of FOXO1 with Lgals9 promoter. Downregulation of Gal-9 enhanced the phosphorylation of ERK, inhibited the expression of p-CREB and IL-10, and promoted the expression of T-bet and IFN-γ in dNK cells. In the mice model, knockout of Lgals9 aggravated adverse pregnancy outcomes caused by T. gondii infection during pregnancy. CONCLUSIONS Toxoplasma gondii infection suppressed Gal-9 expression in dMφ by activating the JNK/FOXO1 signaling pathway, and reduction of Gal-9 contributed to dysfunction of dNK via Gal-9/Tim-3 interaction. This study provides new insights for the molecular mechanisms of the adverse pregnancy outcomes caused by T. gondii.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Shuyan Wang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Xiaoyan Xu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Yuzhu Jiang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Liqin Ren
- Department of Microbiology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Haixia Zhang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Zhidan Li
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Xianbing Liu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| | - Yushan Ren
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong, People's Republic of China.
| |
Collapse
|
14
|
Xie X, Liu J, Gao J, Shang C, Jiang Y, Chen L, Qian Z, Liu L, Wu D, Zhang Y, Ru Z, Zhang Y. The crosstalk between cell death and pregnancy related diseases: A narrative review. Biomed Pharmacother 2024; 176:116815. [PMID: 38788598 DOI: 10.1016/j.biopha.2024.116815] [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: 03/26/2024] [Revised: 05/10/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024] Open
Abstract
Programmed cell death is intricately linked to various physiological phenomena such as growth, development, and metabolism, as well as the proper function of the pancreatic β cell and the migration and invasion of trophoblast cells in the placenta during pregnancy. Traditional and recently identified programmed cell death include apoptosis, autophagy, pyroptosis, necroptosis, and ferroptosis. In addition to cancer and degenerative diseases, abnormal activation of cell death has also been implicated in pregnancy related diseases like preeclampsia, gestational diabetes mellitus, intrahepatic cholestasis of pregnancy, fetal growth restriction, and recurrent miscarriage. Excessive or insufficient cell death and pregnancy related diseases may be mutually determined, ultimately resulting in adverse pregnancy outcomes. In this review, we systematically describe the characteristics and mechanisms underlying several types of cell death and their roles in pregnancy related diseases. Moreover, we discuss potential therapeutic strategies that target cell death signaling pathways for pregnancy related diseases, hoping that more meaningful treatments will be applied in clinical practice in the future.
Collapse
Affiliation(s)
- Xiaowen Xie
- Wuxi Maternal and Child Health Hospital, Wuxi Medical Center of Nanjing Medical University, Wuxi, Jiangsu 214002, China; The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Jiayu Liu
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu 214002, China
| | - Jingyi Gao
- Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chenwei Shang
- Wuxi Maternal and Child Health Hospital, Wuxi Medical Center of Nanjing Medical University, Wuxi, Jiangsu 214002, China; The First Clinical Medical College of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ying Jiang
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu 214002, China
| | - Lingyan Chen
- Wuxi Maternal and Child Health Hospital, Wuxi Medical Center of Nanjing Medical University, Wuxi, Jiangsu 214002, China
| | - Zhiwen Qian
- Wuxi Maternal and Child Health Hospital, Wuxi Medical Center of Nanjing Medical University, Wuxi, Jiangsu 214002, China
| | - Lu Liu
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu 214002, China
| | - Danping Wu
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu 214002, China
| | - Yun Zhang
- Wuxi Maternal and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu 214002, China.
| | - Zhu Ru
- Anqing Medical College Clinical Research Center, Anqing Municipal Hospital, Anqing 246003, Anhui, China.
| | - Yan Zhang
- Wuxi Maternal and Child Health Hospital, Wuxi Medical Center of Nanjing Medical University, Wuxi, Jiangsu 214002, China; Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Affiliated Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu 214002, China.
| |
Collapse
|
15
|
Bezemer RE, Faas MM, van Goor H, Gordijn SJ, Prins JR. Decidual macrophages and Hofbauer cells in fetal growth restriction. Front Immunol 2024; 15:1379537. [PMID: 39007150 PMCID: PMC11239338 DOI: 10.3389/fimmu.2024.1379537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Placental macrophages, which include maternal decidual macrophages and fetal Hofbauer cells, display a high degree of phenotypical and functional plasticity. This provides these macrophages with a key role in immunologically driven events in pregnancy like host defense, establishing and maintaining maternal-fetal tolerance. Moreover, placental macrophages have an important role in placental development, including implantation of the conceptus and remodeling of the intrauterine vasculature. To facilitate these processes, it is crucial that placental macrophages adapt accordingly to the needs of each phase of pregnancy. Dysregulated functionalities of placental macrophages are related to placental malfunctioning and have been associated with several adverse pregnancy outcomes. Although fetal growth restriction is specifically associated with placental insufficiency, knowledge on the role of macrophages in fetal growth restriction remains limited. This review provides an overview of the distinct functionalities of decidual macrophages and Hofbauer cells in each trimester of a healthy pregnancy and aims to elucidate the mechanisms by which placental macrophages could be involved in the pathogenesis of fetal growth restriction. Additionally, potential immune targeted therapies for fetal growth restriction are discussed.
Collapse
Affiliation(s)
- Romy Elisa Bezemer
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Marijke M Faas
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Sanne Jehanne Gordijn
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| | - Jelmer R Prins
- Department of Obstetrics and Gynecology, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
16
|
Jia Y, Huang Y, Ai ZH, Dong YJ, Guo YH, Yang Y, Zhang D, Ye HX, Yang J. Exploring the effectiveness of endometrial receptivity array and immune profiling in patients with multiple implantation failure:A retrospective cohort study based on propensity score matching. J Reprod Immunol 2024; 163:104218. [PMID: 38422808 DOI: 10.1016/j.jri.2024.104218] [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: 10/22/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
This study aimed to evaluate the effectiveness of the endometrial receptivity array (ERA), endometrial immune profiling, and a combination of both in improving the pregnancy outcomes for multiple implantation failure patients. According to patients' willingness, 1429 women who incurred at least two or more consecutive implantation failures in IVF/ICSI treatment opted for frozen embryo transfer and were divided into four groups: 'No test', 'Immune Profiling', 'ERA' and 'ERA+ Immune Profiling'. Women in three test groups underwent timed endometrial biopsy for ERA, immune profiling, a combination of both. We observed the overall incidence rates of the displaced window of implantation (WOI) and endometrial immune dysregulation were 75.14% and 79.29%, respectively. After 1:1 propensity score matching (PSM), our data revealed that the 'ERA' and 'ERA + Immune Profiling' groups demonstrated significantly higher rates of biochemical, clinical, ongoing pregnancy, and implantation compared to the 'No test' group (p < 0.01). The 'Immune Profiling' group showed a higher implantation rate compared to 'No test' group (p < 0.05). Furthermore, when comparing three test groups, the 'ERA + Immune Profiling' group exhibited notably higher rates of clinical and ongoing pregnancy compared to the 'Immune Profiling' group (p < 0.017). However, there was no association between endometrial immune profiling and ERA phases, and their results did not differ between embryo implantation and non-implantation in these patients. Our findings underline the increased implantation rates by use of ERA and endometrial immune profiling in patients with multiple implantation failure, either individually or corporately. Moreover, a combination of both could improve their pregnancy outcomes significantly.
Collapse
Affiliation(s)
- Yan Jia
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, PR China; Department of Reproductive Immunology, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, Sichuan, PR China
| | - Yan Huang
- Department of Reproductive Immunology, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, Sichuan, PR China
| | - Zhong-Hua Ai
- Institute of Health Studies, School of Public Health, Kunming Medical University, Kunming, Yunnan, PR China
| | - Ya-Jun Dong
- Department of Reproductive Immunology, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, Sichuan, PR China
| | - Yan-Hua Guo
- Department of Reproductive Immunology, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, Sichuan, PR China
| | - Yue Yang
- Department of Reproductive Immunology, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, Sichuan, PR China
| | - Dan Zhang
- Department of Reproductive Immunology, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, Sichuan, PR China
| | - Hong-Xia Ye
- Department of Reproductive Immunology, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, Sichuan, PR China; Jinxin Research Institute for Reproductive Medicine and Genetics, Chengdu, Sichuan, PR China.
| | - Jie Yang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, PR China; Department of Reproductive Immunology, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, Sichuan, PR China.
| |
Collapse
|
17
|
Bacon SJ, Zhu Y, Ghosh P. Early spiral arteriole remodeling in the uterine-placental interface: A rat model. J Anat 2024; 244:1054-1066. [PMID: 38288680 PMCID: PMC11095304 DOI: 10.1111/joa.14019] [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: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 05/16/2024] Open
Abstract
The mammalian placenta's interface with the parent is a richly vascularized tissue whose development relies upon communication between many different cell types within the uterine microenvironment. The uterine blood vessels of the interface are reshaped during pregnancy into wide-bore, flaccid vessels that convey parental blood to the exchange region of the placenta. Invasive trophoblast as well as parental uterine macrophages and Natural Killer cells are involved in the stepwise remodeling of these vessels and their respective contributions to this crucial process are still being delineated. However, the earliest steps in arteriole remodeling are understudied as they are difficult to study in humans, and other species lack the deep trophoblast invasion that is so prominent a feature of placentation in humans. Here, we further characterize the rat, with deep hemochorial placentation akin to humans, as a model system in which to tease apart the earliest, relatively understudied events in spiral arteriole remodeling. We show that the rat uterine-placental interface increases in size and vascularity rapidly, before trophoblast invasion. The remodeling stages in the arterioles of the rat uterine-placental interface follow a sequence of anatomical changes similar to those in humans, and there are changes to the arterioles' muscular tunica media prior to the marked influx of immune cells. The rat is a tractable model in which to better understand the cell/cell interactions occurring in vivo in an intact tissue microenvironment over time.
Collapse
Affiliation(s)
- Sarah J Bacon
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Yuxi Zhu
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Priyanjali Ghosh
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| |
Collapse
|
18
|
Wei D, Su Y, Leung PCK, Li Y, Chen ZJ. Roles of bone morphogenetic proteins in endometrial remodeling during the human menstrual cycle and pregnancy. Hum Reprod Update 2024; 30:215-237. [PMID: 38037193 DOI: 10.1093/humupd/dmad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
BACKGROUND During the human menstrual cycle and pregnancy, the endometrium undergoes a series of dynamic remodeling processes to adapt to physiological changes. Insufficient endometrial remodeling, characterized by inadequate endometrial proliferation, decidualization and spiral artery remodeling, is associated with infertility, endometriosis, dysfunctional uterine bleeding, and pregnancy-related complications such as preeclampsia and miscarriage. Bone morphogenetic proteins (BMPs), a subset of the transforming growth factor-β (TGF-β) superfamily, are multifunctional cytokines that regulate diverse cellular activities, such as differentiation, proliferation, apoptosis, and extracellular matrix synthesis, are now understood as integral to multiple reproductive processes in women. Investigations using human biological samples have shown that BMPs are essential for regulating human endometrial remodeling processes, including endometrial proliferation and decidualization. OBJECTIVE AND RATIONALE This review summarizes our current knowledge on the known pathophysiological roles of BMPs and their underlying molecular mechanisms in regulating human endometrial proliferation and decidualization, with the goal of promoting the development of innovative strategies for diagnosing, treating and preventing infertility and adverse pregnancy complications associated with dysregulated human endometrial remodeling. SEARCH METHODS A literature search for original articles published up to June 2023 was conducted in the PubMed, MEDLINE, and Google Scholar databases, identifying studies on the roles of BMPs in endometrial remodeling during the human menstrual cycle and pregnancy. Articles identified were restricted to English language full-text papers. OUTCOMES BMP ligands and receptors and their transduction molecules are expressed in the endometrium and at the maternal-fetal interface. Along with emerging technologies such as tissue microarrays, 3D organoid cultures and advanced single-cell transcriptomics, and given the clinical availability of recombinant human proteins and ongoing pharmaceutical development, it is now clear that BMPs exert multiple roles in regulating human endometrial remodeling and that these biomolecules (and their receptors) can be targeted for diagnostic and therapeutic purposes. Moreover, dysregulation of these ligands, their receptors, or signaling determinants can impact endometrial remodeling, contributing to infertility or pregnancy-related complications (e.g. preeclampsia and miscarriage). WIDER IMPLICATIONS Although further clinical trials are needed, recent advancements in the development of recombinant BMP ligands, synthetic BMP inhibitors, receptor antagonists, BMP ligand sequestration tools, and gene therapies have underscored the BMPs as candidate diagnostic biomarkers and positioned the BMP signaling pathway as a promising therapeutic target for addressing infertility and pregnancy complications related to dysregulated human endometrial remodeling.
Collapse
Affiliation(s)
- Daimin Wei
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Yaxin Su
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Yan Li
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong, China
| |
Collapse
|
19
|
Chen H, Chen Y, Zheng Q. The regulated cell death at the maternal-fetal interface: beneficial or detrimental? Cell Death Discov 2024; 10:100. [PMID: 38409106 PMCID: PMC10897449 DOI: 10.1038/s41420-024-01867-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/08/2024] [Accepted: 02/14/2024] [Indexed: 02/28/2024] Open
Abstract
Regulated cell death (RCD) plays a fundamental role in placental development and tissue homeostasis. Placental development relies upon effective implantation and invasion of the maternal decidua by the trophoblast and an immune tolerant environment maintained by various cells at the maternal-fetal interface. Although cell death in the placenta can affect fetal development and even cause pregnancy-related diseases, accumulating evidence has revealed that several regulated cell death were found at the maternal-fetal interface under physiological or pathological conditions, the exact types of cell death and the precise molecular mechanisms remain elusive. In this review, we summarized the apoptosis, necroptosis and autophagy play both promoting and inhibiting roles in the differentiation, invasion of trophoblast, remodeling of the uterine spiral artery and decidualization, whereas ferroptosis and pyroptosis have adverse effects. RCD serves as a mode of communication between different cells to better maintain the maternal-fetal interface microenvironment. Maintaining the balance of RCD at the maternal-fetal interface is of utmost importance for the development of the placenta, establishment of an immune microenvironment, and prevention of pregnancy disorders. In addition, we also revealed an association between abnormal expression of key molecules in different types of RCD and pregnancy-related diseases, which may yield significant insights into the pathogenesis and treatment of pregnancy-related complications.
Collapse
Affiliation(s)
- Huan Chen
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, P.R. China
| | - Yin Chen
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, P.R. China
| | - Qingliang Zheng
- Prenatal Diagnosis Center, The Eighth Affiliated Hospital, Sun Yat-sen University, 3025# Shennan Road, Shenzhen, 518000, P.R. China.
| |
Collapse
|
20
|
Wu Q, Ying X, Yu W, Li H, Wei W, Lin X, Yang M, Zhang X. Comparison of immune-related gene signatures and immune infiltration features in early- and late-onset preeclampsia. J Gene Med 2024; 26:e3676. [PMID: 38362844 DOI: 10.1002/jgm.3676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/02/2024] [Accepted: 01/28/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Preeclampsia, a severe pregnancy syndrome, is widely accepted divided into early- and late-onset preeclampsia (EOPE and LOPE) based on the onset time of preeclampsia, with distinct pathophysiological origins. However, the molecular mechanism especially immune-related mechanisms for EOPE and LOPE is currently obscure. In the present study, we focused on placental immune alterations between EOPE and LOPE and search for immune-related biomarkers that could potentially serve as potential therapeutic targets through bioinformatic analysis. METHODS The gene expression profiling data was obtained from the Gene Expression Omnibus database. ESTIMATE algorithm and Gene Set Enrichment Analysis were employed to evaluate the immune status. The intersection of differentially expressed genes in GSE74341 series and immune-related genes set screened differentially expressed immune-related genes. Protein-protein interaction network and random forest were used to identify hub genes with a validation by a quantitative real-time PCR. Kyoto Encyclopedia of Genes and Genomes pathways, Gene Ontology and gene set variation analysis were utilized to conduct biological function and pathway enrichment analyses. Single-sample gene set enrichment analysis and CIBERSORTx tools were employed to calculate the immune cell infiltration score. Correlation analyses were evaluated by Pearson correlation analysis. Hub genes-miRNA network was performed by the NetworkAnalyst online tool. RESULTS Immune score and stromal score were all lower in EOPE samples. The immune system-related gene set was significantly downregulated in EOPE compared to LOPE samples. Four hub differentially expressed immune-related genes (IL15, GZMB, IL1B and CXCL12) were identified based on a protein-protein interaction network and random forest. Quantitative real-time polymerase chain reaction validated the lower expression levels of four hub genes in EOPE compared to LOPE samples. Immune cell infiltration analysis found that innate and adaptive immune cells were apparent lacking in EOPE samples compared to LOPE samples. Cytokine-cytokine receptor, para-inflammation, major histocompatibility complex class I and T cell co-stimulation pathways were significantly deficient and highly correlated with hub genes. We constructed a hub genes-miRNA regulatory network, revealing the correlation between hub genes and hsa-miR-374a-5p, hsa-miR-203a-3p, hsa-miR-128-3p, hsa-miR-155-3p, hsa-miR-129-2-3p and hsa-miR-7-5p. CONCLUSIONS The innate and adaptive immune systems were severely impaired in placentas of EOPE. Four immune-related genes (IL15, GZMB, IL1B and CXCL12) were closely correlated with immune-related pathogenesis of EOPE. The result of our study may provide a new basis for discriminating between EOPE and LOPE and acknowledging the role of the immune landscape in the eventual interference and tailored treatment of EOPE.
Collapse
Affiliation(s)
- Quanfeng Wu
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases Xiamen, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen, China
| | - Xiang Ying
- Department of Gynecology and Obstetrics, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Shanghai, China
| | - Weiwei Yu
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Huanxi Li
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Wei Wei
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Xueyan Lin
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Meilin Yang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases Xiamen, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen, China
| | - Xueqin Zhang
- Department of Obstetrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases Xiamen, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen, China
| |
Collapse
|
21
|
Sousa LG, Alves P, Teixeira N, Correia-da-Silva G, Fonseca BM. Alterations in the pro-resolving lipid mediator machinery within first trimester maternal tissue: Implications in decidualization and miscarriage risk. Prostaglandins Leukot Essent Fatty Acids 2024; 201:102619. [PMID: 38788346 DOI: 10.1016/j.plefa.2024.102619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024]
Abstract
A pivotal event in uterine receptivity and human reproduction is the differentiation of endometrial stromal cells into decidual cells, known as decidualization. Decidualization is interlinked with its inflammatory environment. Our study aimed to investigate the presence and role of pro-resolving lipid mediators in first trimester maternal tissue. We assessed the levels of LXA4 and RvD1, along with their metabolic LOX enzymes, in elective (control) and sporadic miscarriage samples. We investigated the effects of LXA4 and RvD1 on decidualization using primary endometrial stromal cells and the immortalized endometrial stromal St-T1b cell line. The upregulation of 12- and 15-LOX expression was observed in pregnancy tissue after sporadic miscarriage, suggesting an inflammatory imbalance. Furthermore, incubation with these lipid mediators led to a decrease in decidualization biomarkers PRL and IGFBP-1, accompanied by morphological changes indicative of aberrant differentiation. The expression of LOX enzymes in decidual natural killer cells suggests their involvement in regulating the inflammatory surroundings and the extent of decidualization.
Collapse
Affiliation(s)
- Luísa G Sousa
- UCIBIO - Applied Molecular Biosciences Unit, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia da Universidade do Porto, 4050-313 Porto, Portugal; i4HB-Institute for Health and Bioeconomy, Universidade do Porto, 4050-313 Porto, Portugal; Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Patrícia Alves
- UCIBIO - Applied Molecular Biosciences Unit, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia da Universidade do Porto, 4050-313 Porto, Portugal; i4HB-Institute for Health and Bioeconomy, Universidade do Porto, 4050-313 Porto, Portugal
| | - Natércia Teixeira
- UCIBIO - Applied Molecular Biosciences Unit, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia da Universidade do Porto, 4050-313 Porto, Portugal; i4HB-Institute for Health and Bioeconomy, Universidade do Porto, 4050-313 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO - Applied Molecular Biosciences Unit, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia da Universidade do Porto, 4050-313 Porto, Portugal; i4HB-Institute for Health and Bioeconomy, Universidade do Porto, 4050-313 Porto, Portugal
| | - Bruno M Fonseca
- UCIBIO - Applied Molecular Biosciences Unit, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Faculdade de Farmácia da Universidade do Porto, 4050-313 Porto, Portugal; i4HB-Institute for Health and Bioeconomy, Universidade do Porto, 4050-313 Porto, Portugal; Escola Superior de Saúde, Instituto Politécnico de Viana do Castelo (IPVC), 4900-347 Viana do Castelo, Portugal.
| |
Collapse
|
22
|
Medegan Fagla B, Buhimschi IA. Protein Misfolding in Pregnancy: Current Insights, Potential Mechanisms, and Implications for the Pathogenesis of Preeclampsia. Molecules 2024; 29:610. [PMID: 38338354 PMCID: PMC10856193 DOI: 10.3390/molecules29030610] [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: 12/18/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Protein misfolding disorders are a group of diseases characterized by supra-physiologic accumulation and aggregation of pathogenic proteoforms resulting from improper protein folding and/or insufficiency in clearance mechanisms. Although these processes have been historically linked to neurodegenerative disorders, such as Alzheimer's disease, evidence linking protein misfolding to other pathologies continues to emerge. Indeed, the deposition of toxic protein aggregates in the form of oligomers or large amyloid fibrils has been linked to type 2 diabetes, various types of cancer, and, in more recent years, to preeclampsia, a life-threatening pregnancy-specific disorder. While extensive physiological mechanisms are in place to maintain proteostasis, processes, such as aging, genetic factors, or environmental stress in the form of hypoxia, nutrient deprivation or xenobiotic exposures can induce failure in these systems. As such, pregnancy, a natural physical state that already places the maternal body under significant physiological stress, creates an environment with a lower threshold for aberrant aggregation. In this review, we set out to discuss current evidence of protein misfolding in pregnancy and potential mechanisms supporting a key role for this process in preeclampsia pathogenesis. Improving our understanding of this emerging pathophysiological process in preeclampsia can lead to vital discoveries that can be harnessed to create better diagnoses and treatment modalities for the disorder.
Collapse
Affiliation(s)
| | - Irina Alexandra Buhimschi
- Department of Obstetrics and Gynecology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| |
Collapse
|
23
|
Yin T, Li X, Li Y, Zang X, Liu L, Du M. Macrophage plasticity and function in cancer and pregnancy. Front Immunol 2024; 14:1333549. [PMID: 38274812 PMCID: PMC10808357 DOI: 10.3389/fimmu.2023.1333549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
As the soil of life, the composition and shaping process of the immune microenvironment of the uterus is worth exploring. Macrophages, indispensable constituents of the innate immune system, are essential mediators of inflammation and tissue remodeling as well. Recent insights into the heterogeneity of macrophage subpopulations have renewed interest in their functional diversity in both physiological and pathological settings. Macrophages display remarkable plasticity and switch from one phenotype to another. Intrinsic plasticity enables tissue macrophages to perform a variety of functions in response to changing tissue contexts, such as cancer and pregnancy. The remarkable diversity and plasticity make macrophages particularly intriguing cells given their dichotomous role in either attacking or protecting tumors and semi-allogeneic fetuses, which of both are characterized functionally by immunomodulation and neovascularization. Here, we reviewed and compared novel perspectives on macrophage biology of these two settings, including origin, phenotype, differentiation, and essential roles in corresponding microenvironments, as informed by recent studies on the heterogeneity of macrophage identity and function, as well as their mechanisms that might offer opportunities for new therapeutic strategies on malignancy and pregnancy complications.
Collapse
Affiliation(s)
- Tingxuan Yin
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xinyi Li
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yanhong Li
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Lu Liu
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Meirong Du
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
24
|
Vishnyakova P, Gantsova E, Kiseleva V, Lazarev D, Knyazev E, Poltavets A, Iskusnykh M, Muminova K, Potapova A, Khodzhaeva Z, Elchaninov A, Fatkhudinov T, Sukhikh G. MicroRNA miR-27a as a possible regulator of anti-inflammatory macrophage phenotype in preeclamptic placenta. Placenta 2024; 145:151-161. [PMID: 38141416 DOI: 10.1016/j.placenta.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/23/2023] [Accepted: 12/03/2023] [Indexed: 12/25/2023]
Abstract
INTRODUCTION The role of the TGFβ signaling pathway, an important cascade responsible for the anti-inflammatory polarization of macrophages, in the development of both early- and late-onset preeclampsia (eoPE and loPE), remains poorly understood. In this study, we examined the components of the TGFβ signaling cascade and macrophage markers within placental tissue in normal pregnancy and in PE. METHODS Patients with eoPE, loPE, and normal pregnancy were enrolled in the study (n = 10 in each group). Following techniques were used for the investigation: immunohistochemistry analysis, western blotting, qRT-PCR, isolation of monocytes by magnetic sorting, transfection, microRNA sequencing, and bioinformatic analysis. RESULTS We observed a significant decrease in the anti-inflammatory macrophage marker CD206 in the loPE group, alongside with a significant down-regulation of CD206 protein production in both eoPE and loPE groups. The level of CD68-positive cells and relative levels of CD163 and MARCO production were comparable across the groups. However, we identified a significant decrease in the TGFβ receptor 2 production and its gene expression in the PE group. Further analysis revealed a link between TGFBR2 and MRC1 (CD206) genes through a single miRNA, hsa-miR-27a-3p. Transfecting CD14-derived macrophages with the hsa-miR-27a-3p mimic significantly changed TGFBR2 production, indicating the potential role of this miRNA in regulating the TGFβ signaling pathway. We also revealed the up-regulation of hsa-miR-27a-5p and hsa-miR-27a-3p in the trophoblast BeWo b30 cell line under the severe hypoxia condition and the fact that TGFBR2 3' UTR could serve as a potential target for these miRNAs. DISCUSSION Our findings uncover a novel potential therapeutic target for managing patients with PE, significantly contributing to a deeper comprehension of the underlying mechanisms involved in the development of this pathology.
Collapse
Affiliation(s)
- Polina Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia, Moscow, Russia.
| | - Elena Gantsova
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia, Moscow, Russia
| | - Viktoriia Kiseleva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia, Moscow, Russia
| | - Dmitry Lazarev
- Pirogov Russian National Research Medical University (Pirogov Medical University), Moscow, Russia
| | - Evgeny Knyazev
- Faculty of Biology and Biotechnology, HSE University, Moscow, Russia; Laboratory of Microfluidic Technologies for Biomedicine, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Anastasiya Poltavets
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Marina Iskusnykh
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia, Moscow, Russia
| | - Kamilla Muminova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Alena Potapova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Zulfiya Khodzhaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Andrey Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia; Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia, Moscow, Russia; Pirogov Russian National Research Medical University (Pirogov Medical University), Moscow, Russia; Avtsyn Research Institute of Human Morphology of Federal state budgetary scientific institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
| | - Timur Fatkhudinov
- Research Institute of Molecular and Cellular Medicine, Peoples' Friendship University of Russia, Moscow, Russia; Avtsyn Research Institute of Human Morphology of Federal state budgetary scientific institution "Petrovsky National Research Centre of Surgery", Moscow, Russia
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| |
Collapse
|
25
|
Hu M, Zhang Y, Zhang X, Zhang X, Huang X, Lu Y, Li Y, Brännström M, Sferruzzi-Perri AN, Shao LR, Billig H. Defective Uterine Spiral Artery Remodeling and Placental Senescence in a Pregnant Rat Model of Polycystic Ovary Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1916-1935. [PMID: 37689383 DOI: 10.1016/j.ajpath.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/28/2023] [Accepted: 08/23/2023] [Indexed: 09/11/2023]
Abstract
Pregnancy-related problems have been linked to impairments in maternal uterine spiral artery (SpA) remodeling. The mechanisms underlying this association are still unclear. It is also unclear whether hyperandrogenism and insulin resistance, the two common manifestations of polycystic ovary syndrome, affect uterine SpA remodeling. We verified previous work in which exposure to 5-dihydrotestosterone (DHT) and insulin (INS) in rats during pregnancy resulted in hyperandrogenism, insulin intolerance, and higher fetal mortality. Exposure to DHT and INS dysregulated the expression of angiogenesis-related genes in the uterus and placenta and also decreased expression of endothelial nitric oxide synthase and matrix metallopeptidases 2 and 9, increased fibrotic collagen deposits in the uterus, and reduced expression of marker genes for SpA-associated trophoblast giant cells. These changes were related to a greater proportion of unremodeled uterine SpAs and a smaller proportion of highly remodeled arteries in DHT + INS-exposed rats. Placentas from DHT + INS-exposed rats exhibited decreased basal and labyrinth zone regions, reduced maternal blood spaces, diminished labyrinth vascularity, and an imbalance in the abundance of vascular and smooth muscle proteins. Furthermore, placentas from DHT + INS-exposed rats showed expression of placental insufficiency markers and a significant increase in cell senescence-associated protein levels. Altogether, this work demonstrates that increased pregnancy complications in polycystic ovary syndrome may be mediated by problems with uterine SpA remodeling, placental functionality, and placental senescence.
Collapse
Affiliation(s)
- Min Hu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China; Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yuehui Zhang
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xu Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - XiuYing Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyue Huang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yaxing Lu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yijia Li
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Linus R Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
26
|
Parks SE, Geng T, Monsivais D. Endometrial TGFβ signaling fosters early pregnancy development by remodeling the fetomaternal interface. Am J Reprod Immunol 2023; 90:e13789. [PMID: 38009061 PMCID: PMC10683870 DOI: 10.1111/aji.13789] [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: 05/02/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 11/28/2023] Open
Abstract
The endometrium is a unique and highly regenerative tissue with crucial roles during the reproductive lifespan of a woman. As the first site of contact between mother and embryo, the endometrium, and its critical processes of decidualization and immune cell recruitment, play a leading role in the establishment of pregnancy, embryonic development, and reproductive capacity. These integral processes are achieved by the concerted actions of steroid hormones and a myriad of growth factor signaling pathways. This review focuses on the roles of the transforming growth factor β (TGFβ) pathway in the endometrium during the earliest stages of pregnancy through the lens of immune cell regulation and function. We discuss how key ligands in the TGFβ family signal through downstream SMAD transcription factors and ultimately remodel the endometrium into a state suitable for embryo implantation and development. We also focus on the key roles of the TGFβ signaling pathway in recruiting uterine natural killer cells and their collective remodeling of the decidua and spiral arteries. By providing key details about immune cell populations and TGFβ signaling within the endometrium, it is our goal to shed light on the intricate remodeling that is required to achieve a successful pregnancy.
Collapse
Affiliation(s)
- Sydney E. Parks
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
- Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ting Geng
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
| | - Diana Monsivais
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Drug Discovery, Baylor College of Medicine, Houston, TX 77030, USA
- Cancer and Cell Biology Program, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
27
|
Yokouchi-Konishi T, Liu Y, Feng L. Progesterone receptor membrane component 2 is critical for human placental extravillous trophoblast invasion. Biol Reprod 2023; 109:759-771. [PMID: 37665239 DOI: 10.1093/biolre/ioad109] [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: 03/07/2023] [Revised: 08/14/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023] Open
Abstract
Proper extravillous trophoblast invasion is essential for normal placentation and pregnancy. However, the molecular mechanisms by which cytotrophoblasts differentiate into extravillous trophoblast are unclear. We discovered that in the first-trimester placenta, progesterone receptor membrane component 2 was highly expressed in syncytiotrophoblast but significantly lower in extravillous trophoblast and cytotrophoblasts, indicating a divergent role for progesterone receptor membrane component 2 in trophoblast functions. We aim to examine the role of progesterone receptor membrane component 2 in extravillous trophoblasts invasion mediated by both intracellular and extracellular signals. Progesterone receptor membrane component 2 knockdown and overexpression cells were established in HTR8/SVneo cells, a first-trimester extravillous trophoblast-derived cell model, by transfection with small-interfering RNA or progesterone receptor membrane component 2 plasmids, respectively. Progesterone receptor membrane component 2 knockdown led to cellular morphological changes , enhanced trophoblast proliferation,invasion, and promoted tube formation. These effects were mediated by the activation of hypoxia-inducible factor 1alpha and an increased expression of vascular endothelial growth factor A. The culture supernatant collected from progesterone receptor membrane component 2 knockdown cells did not significantly affect extravillous trophoblast invasion compared to the controls, indicating that extracellular signaling did not robustly regulate extravillous trophoblast invasion in this study. In conclusion, attenuation of progesterone receptor membrane component 2 plays a role in placentation by promoting cell proliferation, invasion, and angiogenesis in extravillous trophoblasts via activation of hypoxia-inducible factor 1 alpha signaling. We thus identified a new function of progesterone receptor membrane component 2 and provide insights on understanding the mechanisms of trophoblast invasion.
Collapse
Affiliation(s)
- Tae Yokouchi-Konishi
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
- Department of Obstetrics and Gynecology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yongjie Liu
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
28
|
Abstract
Although historically pre-eclampsia, preterm birth, abruption, fetal growth restriction and stillbirth have been viewed as clinically distinct entities, a growing body of literature has demonstrated that the placenta and its development is the root cause of many cases of these conditions. This has led to the term 'the great obstetrical syndromes' being coined to reflect this common origin. Although these conditions mostly manifest in the second half of pregnancy, a failure to complete deep placentation (the transition from histiotrophic placentation to haemochorial placenta at 10-18 weeks of gestation via a second wave of extravillous trophoblast invasion), is understood to be key to the pathogenesis of the great obstetrical syndromes. While the reasons that the placenta fails to achieve deep placentation remain active areas of investigation, maternal inflammation and thrombosis have been clearly implicated. From a clinical standpoint these mechanisms provide a biological explanation of how low-dose aspirin, which affects the COX-1 receptor (thrombosis) and the COX-2 receptor (inflammation), prevents not just pre-eclampsia but all the components of the great obstetrical syndromes if initiated early in pregnancy. The optimal dose of low-dose aspirin that is maximally effective in pregnancy remains a question open for further research. Additionally, other candidate medications have been identified that may also prevent pre-eclampsia, and further study of them may offer therapeutic options beyond low-dose aspirin. Interestingly, three of the eight identified compounds (hydroxychloroquine, metformin and pravastatin) are known to decrease inflammation.
Collapse
Affiliation(s)
- Matthew K Hoffman
- Departments of Obstetrics and Gynecology, Christiana Care Health Services, Newark, Delaware, USA
| |
Collapse
|
29
|
Olaya-C M, Franco Zuluaga JA. More Tools for Evaluating Decidual Artery Disease. Int J Surg Pathol 2023; 31:1217-1224. [PMID: 36471503 DOI: 10.1177/10668969221140386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Introduction: Hypertensive disorders of pregnancy continue to pose the most important risks for adverse maternal and neonatal outcome. Among histological findings, decidual artery disease is one of the most common, one that has both good reproducibility among observers and whose abnormal vascular remodeling is the sole aspect of preeclampsia pathophysiology on which experts agree. Nevertheless, some aspects of arterial remodeling alterations are still under investigation. Methods: We selected 720 routine and consecutive placenta case studies, concordant with the Amsterdam consensus. From these studies, we collected maternal and neonatal clinical data and specific placental findings on spiral artery abnormalities. We took into account all criteria for decidual arteriopathy. Two hundred and fifteen (215) cases out of this population presented hypertensive disorders of pregnancy. Additional to expected arterial findings, we noted frequent persistent parietal trophoblast lining. Results: A large proportion of our population developed hypertensive disorders of pregnancy (30%). Among the histologic findings reported for preeclampsia, we paid particular attention to spiral artery abnormalities, and this interpretive analysis revealed high frequency of arterial remodeling abnormalities. We examined two additional aspects in our routine analysis: first, the novel one of parietal trophoblast persistence, and second, the established problem of associated acute inflammation, as a possible pitfall. Conclusion: In order to better understood, spiral maternal artery remodeling merits further study. The abnormalities in this process provide an objective tool in the study and diagnosis of important pregnancy complications; furthermore, abnormal remodeling is an expression of early pregnancy alteration, and subsequently related to preeclampsia etiology.
Collapse
Affiliation(s)
- Mercedes Olaya-C
- Department of Pathology, Institute of Human Genetics, the Medical School, Pontificia Universidad Javeriana, Hospital Universitario San Ignacio, Bogota, Colombia
| | - Jorge A Franco Zuluaga
- Department of Morphology, the Medical School, Pontificia Universidad Javeriana, Bogota, Colombia
| |
Collapse
|
30
|
Zhang L, Liu J, Feng X, Lash GE. Unraveling the mysteries of spiral artery remodeling. Placenta 2023; 141:51-56. [PMID: 37308346 DOI: 10.1016/j.placenta.2023.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/11/2023] [Accepted: 05/17/2023] [Indexed: 06/14/2023]
Abstract
Spiral artery remodeling is the process by which the uterine vessels become large bore low resistance conduits, allowing delivery of high volumes of maternal blood to the placenta to nourish the developing fetus. Failure of this process is associated with the pathophysiology of most of the major obstetric complications, including late miscarriage, fetal growth restriction and pre-eclampsia. However, the point at which remodeling 'fails' in these pathological pregnancies is not yet clear. Spiral artery remodeling has predominantly been described in terms of its morphological features, however we are starting to understand more about the cellular and molecular triggers of the different aspects of this process. This review will discuss the current state of knowledge of spiral artery remodeling, in particular the processes involved in loss of the vascular smooth muscle cells, and consider where in the process defects would lead to a pathological pregnancy.
Collapse
Affiliation(s)
- Lindong Zhang
- Department of Gynecology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Liu
- Department of Gynecology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoqian Feng
- Department of Gynecology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gendie E Lash
- Department of Gynecology, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Division of Uterine Vascular Biology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China.
| |
Collapse
|
31
|
Gualdoni GS, Barril C, Jacobo PV, Pacheco Rodríguez LN, Cebral E. Involvement of metalloproteinase and nitric oxide synthase/nitric oxide mechanisms in early decidual angiogenesis-vascularization of normal and experimental pathological mouse placenta related to maternal alcohol exposure. Front Cell Dev Biol 2023; 11:1207671. [PMID: 37670932 PMCID: PMC10476144 DOI: 10.3389/fcell.2023.1207671] [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: 04/17/2023] [Accepted: 07/28/2023] [Indexed: 09/07/2023] Open
Abstract
Successful pregnancy for optimal fetal growth requires adequate early angiogenesis and remodeling of decidual spiral arterioles during placentation. Prior to the initiation of invasion and endothelial replacement by trophoblasts, interactions between decidual stromal cells and maternal leukocytes, such as uterine natural killer cells and macrophages, play crucial roles in the processes of early maternal vascularization, such as proliferation, apoptosis, migration, differentiation, and matrix and vessel remodeling. These placental angiogenic events are highly dependent on the coordination of several mechanisms at the early maternal-fetal interface, and one of them is the expression and activity of matrix metalloproteinases (MMPs) and endothelial nitric oxide synthases (NOSs). Inadequate balances of MMPs and nitric oxide (NO) are involved in several placentopathies and pregnancy complications. Since alcohol consumption during gestation can affect fetal growth associated with abnormal placental development, recently, we showed, in a mouse model, that perigestational alcohol consumption up to organogenesis induces fetal malformations related to deficient growth and vascular morphogenesis of the placenta at term. In this review, we summarize the current knowledge of the early processes of maternal vascularization that lead to the formation of the definitive placenta and the roles of angiogenic MMP and NOS/NO mechanisms during normal and altered early gestation in mice. Then, we propose hypothetical defective decidual cellular and MMP and NOS/NO mechanisms involved in abnormal decidual vascularization induced by perigestational alcohol consumption in an experimental mouse model. This review highlights the important roles of decidual cells and their MMP and NOS balances in the physiological and pathophysiological early maternal angiogenesis-vascularization during placentation in mice.
Collapse
Affiliation(s)
| | | | | | | | - Elisa Cebral
- Laboratorio de Reproducción y Fisiología Materno-Embrionaria, Instituto de Biodiversidad y Biología Experimental y Aplicada (IBBEA), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET), Departamento de Biodiversidad y Biología Experimental (DBBE), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
32
|
Lawless L, Qin Y, Xie L, Zhang K. Trophoblast Differentiation: Mechanisms and Implications for Pregnancy Complications. Nutrients 2023; 15:3564. [PMID: 37630754 PMCID: PMC10459728 DOI: 10.3390/nu15163564] [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: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Placental development is a tightly controlled event, in which cell expansion from the trophectoderm occurs in a spatiotemporal manner. Proper trophoblast differentiation is crucial to the vitality of this gestational organ. Obstructions to its development can lead to pregnancy complications, such as preeclampsia, fetal growth restriction, and preterm birth, posing severe health risks to both the mother and offspring. Currently, the only known treatment strategy for these complications is delivery, making it an important area of research. The aim of this review was to summarize the known information on the development and mechanistic regulation of trophoblast differentiation and highlight the similarities in these processes between the human and mouse placenta. Additionally, the known biomarkers for each cell type were compiled to aid in the analysis of sequencing technologies.
Collapse
Affiliation(s)
- Lauren Lawless
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX 77030, USA;
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Yushu Qin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Ke Zhang
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX 77030, USA;
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
33
|
Guterstam YC, Acharya G, Schott K, Björkström NK, Gidlöf S, Ivarsson MA. Immune cell profiling of vaginal blood from patients with early pregnancy bleeding. Am J Reprod Immunol 2023; 90:e13738. [PMID: 37491928 DOI: 10.1111/aji.13738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/20/2023] [Accepted: 06/12/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM Vaginal bleeding during early pregnancy is estimated to occur in 20% of all pregnancies and it is often difficult to predict who will ultimately miscarry. The role of immune cells in early pregnancy loss is poorly understood. METHOD OF STUDY In this prospective cohort study, 28 pregnant women presenting with first-trimester vaginal bleeding donated vaginal blood, peripheral venous blood, and saliva during their initial emergency room visit, and at a follow-up. The composition, frequency, and phenotype of immune cells in the vaginal blood were determined using flow cytometry. The proteome of serum and saliva was analyzed with OLINK proximity extension assay and correlated to vaginal immune cell phenotype and outcome of pregnancy. The course and outcome of pregnancies were followed and recorded. RESULTS Vaginal blood contained all main immune cell lineages including B cells, NK cells, T cells, and monocytes/macrophages. Notably, vaginal blood immune cells expressed tissue residency markers including CD49a. Women who subsequently miscarried had a higher frequency of vaginal blood CD49a+ NK cells compared to those who did not miscarry, and this correlated with serum levels of granzyme A and H, as well as CSF1, CAIX, and TWEAK. Women in the miscarriage group also had a higher frequency of peripheral blood T cells expressing CD49a. CONCLUSIONS Our study provides novel insight into human reproductive immunology in relation to miscarriage. Tissue-resident NK cells in vaginal blood alone or in combination with serological biomarkers hold potential as prognostic factors in the prediction of pregnancy outcome in women with early pregnancy bleedings.
Collapse
Affiliation(s)
- Ylva Crona Guterstam
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Ganesh Acharya
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Center for Fetal Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Katharina Schott
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sebastian Gidlöf
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Martin A Ivarsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
34
|
Doroftei B, Ilie OD, Armeanu T, Stoian IL, Anton N, Babici RG, Ilea C. A Narrative Review Discussing the Obstetric Repercussions Due to Alterations of Personalized Bacterial Sites Developed within the Vagina, Cervix, and Endometrium. J Clin Med 2023; 12:5069. [PMID: 37568471 PMCID: PMC10419759 DOI: 10.3390/jcm12155069] [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: 05/31/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND The reproductive tract microbiota that evolved as an integrative component has been studied intensively in the last decade. As a result, novel research, clinical opportunities, and perspectives have been derived following the close investigation of this microecological environment. This has paved the way for an update to and improvement of the management strategies and therapeutic approaches. However, obscurities, contradictions, and controversies arise regarding the ascension route from the vagina to the endometrium via the cervix, with finality in adverse obstetric outcomes. METHODS Starting from these considerations, we aimed to gather all existing data and information from four major academic databases (PubMed, ISI Web of Knowledge, Scopus, and ScienceDirect) published in the last 13 years (2010-2023) using a controlled vocabulary and dedicated terminology to enhance the coverage, identification, and sorting of potentially eligible studies. RESULTS Despite the high number of returned entries (n = 804), only a slight percentage (2.73%) of all manuscripts were deemed eligible following two rounds of evaluation. Cumulatively, a low level of Lactobacillus spp. and of other core microbiota members is mandatory, with a possible eubiosis-to-dysbiosis transition leading to an impairment of metabolic and endocrine network homeostasis. This transposes into a change in the pro-inflammatory landscape and activation of signaling pathways due to activity exerted by the bacterial lipopolysaccharides (LPSs)/endotoxins that further reflect a high risk of miscarriage in various stages. While the presence of some pathogenic entities may be suggestive of an adverse obstetric predisposition, there are still pros and cons of the role of specific strains, as only the vagina and cervix have been targeted as opposed to the endometrium, which recently started to be viewed as the key player in the vagina-cervix-endometrium route. Consequently, based on an individual's profile, diet, and regime, antibiotics and probiotics might be practical or not. CONCLUSIONS Resident bacteria have a dual facet and are beneficial for women's health, but, at the same time, relaying on the abundance, richness, and evenness that are definitory indexes standing as intermediaries of a miscarriage.
Collapse
Affiliation(s)
- Bogdan Doroftei
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street No. 34, 700038 Iasi, Romania
- Origyn Fertility Center, Palace Street No. 3C, 700032 Iasi, Romania
| | | | - Theodora Armeanu
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street No. 34, 700038 Iasi, Romania
- Origyn Fertility Center, Palace Street No. 3C, 700032 Iasi, Romania
| | - Irina-Liviana Stoian
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
| | - Nicoleta Anton
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
| | - Ramona-Geanina Babici
- Department of Genetics, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
| | - Ciprian Ilea
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania
- Clinical Hospital of Obstetrics and Gynecology “Cuza Voda”, Cuza Voda Street No. 34, 700038 Iasi, Romania
| |
Collapse
|
35
|
Greenbaum S, Averbukh I, Soon E, Rizzuto G, Baranski A, Greenwald NF, Kagel A, Bosse M, Jaswa EG, Khair Z, Kwok S, Warshawsky S, Piyadasa H, Goldston M, Spence A, Miller G, Schwartz M, Graf W, Van Valen D, Winn VD, Hollmann T, Keren L, van de Rijn M, Angelo M. A spatially resolved timeline of the human maternal-fetal interface. Nature 2023; 619:595-605. [PMID: 37468587 PMCID: PMC10356615 DOI: 10.1038/s41586-023-06298-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 06/08/2023] [Indexed: 07/21/2023]
Abstract
Beginning in the first trimester, fetally derived extravillous trophoblasts (EVTs) invade the uterus and remodel its spiral arteries, transforming them into large, dilated blood vessels. Several mechanisms have been proposed to explain how EVTs coordinate with the maternal decidua to promote a tissue microenvironment conducive to spiral artery remodelling (SAR)1-3. However, it remains a matter of debate regarding which immune and stromal cells participate in these interactions and how this evolves with respect to gestational age. Here we used a multiomics approach, combining the strengths of spatial proteomics and transcriptomics, to construct a spatiotemporal atlas of the human maternal-fetal interface in the first half of pregnancy. We used multiplexed ion beam imaging by time-of-flight and a 37-plex antibody panel to analyse around 500,000 cells and 588 arteries within intact decidua from 66 individuals between 6 and 20 weeks of gestation, integrating this dataset with co-registered transcriptomics profiles. Gestational age substantially influenced the frequency of maternal immune and stromal cells, with tolerogenic subsets expressing CD206, CD163, TIM-3, galectin-9 and IDO-1 becoming increasingly enriched and colocalized at later time points. By contrast, SAR progression preferentially correlated with EVT invasion and was transcriptionally defined by 78 gene ontology pathways exhibiting distinct monotonic and biphasic trends. Last, we developed an integrated model of SAR whereby invasion is accompanied by the upregulation of pro-angiogenic, immunoregulatory EVT programmes that promote interactions with the vascular endothelium while avoiding the activation of maternal immune cells.
Collapse
Affiliation(s)
- Shirley Greenbaum
- Department of Pathology, Stanford University, Stanford, CA, USA.
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
| | - Inna Averbukh
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Erin Soon
- Department of Pathology, Stanford University, Stanford, CA, USA
- Immunology Program, Stanford University, Stanford, CA, USA
| | - Gabrielle Rizzuto
- Department of Pathology, University of Californica San Francisco, San Francisco, CA, USA
| | - Alex Baranski
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Noah F Greenwald
- Department of Pathology, Stanford University, Stanford, CA, USA
- Cancer Biology Program, Stanford University, Stanford, CA, USA
| | - Adam Kagel
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Marc Bosse
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Eleni G Jaswa
- Department of Obstetrics Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Zumana Khair
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Shirley Kwok
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | | | - Mako Goldston
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Angie Spence
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Geneva Miller
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Morgan Schwartz
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Will Graf
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - David Van Valen
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| | - Travis Hollmann
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Leeat Keren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Michael Angelo
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
36
|
Kanter J, Gordon SM, Mani S, Sokalska A, Park JY, Senapati S, Huh DD, Mainigi M. Hormonal stimulation reduces numbers and impairs function of human uterine natural killer cells during implantation. Hum Reprod 2023; 38:1047-1059. [PMID: 37075311 PMCID: PMC10501469 DOI: 10.1093/humrep/dead069] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/05/2023] [Indexed: 04/21/2023] Open
Abstract
STUDY QUESTION How does an altered maternal hormonal environment, such as that seen during superovulation with gonadotropins in ART, impact human uterine immune cell distribution and function during the window of implantation? SUMMARY ANSWER Hormonal stimulation with gonadotropins alters abundance of maternal immune cells including uterine natural killer (uNK) cells and reduces uNK cell ability to promote extravillous trophoblast (EVT) invasion. WHAT IS KNOWN ALREADY An altered maternal hormonal environment, seen following ART, can lead to increased risk for adverse perinatal outcomes associated with disordered placentation. Maternal immune cells play an essential role in invasion of EVTs, a process required for proper establishment of the placenta, and adverse perinatal outcomes have been associated with altered immune cell populations. How ART impacts maternal immune cells and whether this can in turn affect implantation and placentation in humans remain unknown. STUDY DESIGN, SIZE, DURATION A prospective cohort study was carried out between 2018 and 2021 on 51 subjects: 20 from natural cycles 8 days after LH surge; and 31 from stimulated IVF cycles 7 days after egg retrieval. PARTICIPANTS/MATERIALS, SETTING, METHODS Endometrial biopsies and peripheral blood samples were collected during the window of implantation in subjects with regular menstrual cycles or undergoing superovulation. Serum estradiol and progesterone levels were measured by chemiluminescent competitive immunoassay. Immune cell populations in blood and endometrium were analyzed using flow cytometry. uNK cells were purified using fluorescence-activated cell sorting and were subjected to RNA sequencing (RNA-seq). Functional changes in uNK cells due to hormonal stimulation were evaluated using the implantation-on-a-chip (IOC) device, a novel bioengineered platform using human primary cells that mimics early processes that occur during pregnancy in a physiologically relevant manner. Unpaired t-tests, one-way ANOVA, and pairwise multiple comparison tests were used to statistically evaluate differences. MAIN RESULTS AND THE ROLE OF CHANCE Baseline characteristics were comparable for both groups. As expected, serum estradiol levels on the day of biopsy were significantly higher in stimulated (superovulated) patients (P = 0.0005). In the setting of superovulation, we found an endometrium-specific reduction in the density of bulk CD56+ uNK cells (P < 0.05), as well as in the uNK3 subpopulation (P = 0.025) specifically (CD103+ NK cells). In stimulated samples, we also found that the proportion of endometrial B cells was increased (P < 0.0001). Our findings were specific to the endometrium and not seen in peripheral blood. On the IOC device, uNK cells from naturally cycling secretory endometrium promote EVT invasion (P = 0.03). However, uNK cells from hormonally stimulated endometrium were unable to significantly promote EVT invasion, as measured by area of invasion, depth of invasion, and number of invaded EVTs by area. Bulk RNA-seq of sorted uNK cells from stimulated and unstimulated endometrium revealed changes in signaling pathways associated with immune cell trafficking/movement and inflammation. LIMITATIONS, REASONS FOR CAUTION Patient numbers utilized for the study were low but were enough to identify significant overall population differences in select immune cell types. With additional power and deeper immune phenotyping, we may detect additional differences in immune cell composition of blood and endometrium in the setting of hormonal stimulation. Flow cytometry was performed on targeted immune cell populations that have shown involvement in early pregnancy. A more unbiased approach might identify changes in novel maternal immune cells not investigated in this study. We performed RNA-seq only on uNK cells, which demonstrated differences in gene expression. Ovarian stimulation may also impact gene expression and function of other subsets of immune cells, as well as other cell types within the endometrium. Finally, the IOC device, while a major improvement over existing in vitro methods to study early pregnancy, does not include all possible maternal cells present during early pregnancy, which could impact functional effects seen. Immune cells other than uNK cells may impact invasion of EVTs in vitro and in vivo, though these remain to be tested. WIDER IMPLICATIONS OF THE FINDINGS These findings demonstrate that hormonal stimulation affects the distribution of uNK cells during the implantation window and reduces the proinvasive effects of uNK cells during early pregnancy. Our results provide a potential mechanism by which fresh IVF cycles may increase risk of disorders of placentation, previously linked to adverse perinatal outcomes. STUDY FUNDING/COMPETING INTEREST(S) Research reported in this publication was supported by the University of Pennsylvania University Research Funding (to M.M.), the Eunice Kennedy Shriver National Institute of Child Health and Human Development (P50HD068157 to M.M., S.S., and S.M.), National Center for Advancing Translational Sciences of the National Institutes of Health (TL1TR001880 to J.K.), the Institute for Translational Medicine and Therapeutics of the Perelman School of Medicine at the University of Pennsylvania, the Children's Hospital of Philadelphia Research Institute (to S.M.G.), and the National Institute of Allergy and Infectious Diseases (K08AI151265 to S.M.G.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- J Kanter
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - S M Gordon
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - S Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - A Sokalska
- Division of Reproductive Endocrinology and Infertility, Stanford University, Stanford, CA, USA
| | - J Y Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - S Senapati
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - D D Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - M Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
37
|
Huang CC, Hsueh YW, Chang CW, Hsu HC, Yang TC, Lin WC, Chang HM. Establishment of the fetal-maternal interface: developmental events in human implantation and placentation. Front Cell Dev Biol 2023; 11:1200330. [PMID: 37266451 PMCID: PMC10230101 DOI: 10.3389/fcell.2023.1200330] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Early pregnancy is a complex and well-orchestrated differentiation process that involves all the cellular elements of the fetal-maternal interface. Aberrant trophoblast-decidual interactions can lead to miscarriage and disorders that occur later in pregnancy, including preeclampsia, intrauterine fetal growth restriction, and preterm labor. A great deal of research on the regulation of implantation and placentation has been performed in a wide range of species. However, there is significant species variation regarding trophoblast differentiation as well as decidual-specific gene expression and regulation. Most of the relevant information has been obtained from studies using mouse models. A comprehensive understanding of the physiology and pathology of human implantation and placentation has only recently been obtained because of emerging advanced technologies. With the derivation of human trophoblast stem cells, 3D-organoid cultures, and single-cell analyses of differentiated cells, cell type-specific transcript profiles and functions were generated, and each exhibited a unique signature. Additionally, through integrative transcriptomic information, researchers can uncover the cellular dysfunction of embryonic and placental cells in peri-implantation embryos and the early pathological placenta. In fact, the clinical utility of fetal-maternal cellular trafficking has been applied for the noninvasive prenatal diagnosis of aneuploidies and the prediction of pregnancy complications. Furthermore, recent studies have proposed a viable path toward the development of therapeutic strategies targeting placenta-enriched molecules for placental dysfunction and diseases.
Collapse
|
38
|
Habets DHJ, Al-Nasiry S, Nagelkerke SQ, Voorter CEM, Spaanderman MEA, Kuijpers TW, Wieten L. Analysis of FCGR3A-p.176Val variants in women with recurrent pregnancy loss and the association with CD16a expression and anti-HLA antibody status. Sci Rep 2023; 13:5232. [PMID: 36997584 PMCID: PMC10063683 DOI: 10.1038/s41598-023-32156-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 03/23/2023] [Indexed: 04/01/2023] Open
Abstract
AbstractNatural Killer (NK) cells have been implicated in recurrent pregnancy loss (RPL). The p.Val176Phe (or Val158Phe) Single Nucleotide Polymorphism (SNP) in the FCGR3A gene encoding the FcγRIIIA or CD16a receptor has been associated with an enhanced affinity for IgG and stronger NK-mediated antibody-dependent cellular cytotoxicity. We hypothesized that the presence of at least one p.176Val variant associates with RPL and increased CD16a expression and alloantibodies e.g., against paternal human leukocyte antigen (HLA). In 50 women with RPL, we studied frequencies of the p.Val176Phe FCGR3A polymorphisms. Additionally, CD16a expression and anti-HLA antibody status were analyzed by flowcytometry and Luminex Single Antigens. In woman with RPL, frequencies were: 20% (VV), 42% (VF) and 38% (FF). This was comparable to frequencies from the European population in the NCBI SNP database and in an independent Dutch cohort of healthy women. NK cells from RPL women with a VV (22,575 [18731-24607]) and VF (24,294 [20157-26637]) polymorphism showed a higher expression of the CD16a receptor than NK cells from RPL women with FF (17,367 [13257-19730]). No difference in frequencies of the FCGR3A-p.176 SNP were detected when comparing women with or without class I and class II anti-HLA antibodies. Our study does not provide strong evidence for an association between the p.Val176Phe FCGR3A SNP and RPL.
Collapse
|
39
|
Ismail NI. Relative expression of receptors in uterine natural killer cells compared to peripheral blood natural killer cells. Front Immunol 2023; 14:1166451. [PMID: 37051244 PMCID: PMC10083503 DOI: 10.3389/fimmu.2023.1166451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
One would expect maternal immune cells to attack the invading trophoblast as the placenta is semi-allogenic. However, they appear to cooperate with the trophoblast in disrupting the arterial wall which has been determined in several studies. uNK cells are a particular type of immune cell that appears to play a role in pregnancy. As in pregnancy, the key contributors to trophoblast invasion appear to be a unique combination of genes, which appear to regulate multiple components of the interactions between placental and maternal cells, called HLA class 1b genes. The HLA class 1b genes have few alleles, which makes them unlikely to be recognized as foreign by the maternal cells. The low polymorphic properties of these particular HLAs may aid trophoblasts in actively avoiding immune attacks. This review gives a complete description of the mechanisms of interaction between HLAs and maternal uNK cells in humans.
Collapse
|
40
|
Krop J, Tian X, van der Hoorn ML, Eikmans M. The Mac Is Back: The Role of Macrophages in Human Healthy and Complicated Pregnancies. Int J Mol Sci 2023; 24:5300. [PMID: 36982375 PMCID: PMC10049527 DOI: 10.3390/ijms24065300] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Pregnancy is a fascinating immunological paradox: the semi-allogeneic fetus generally grows without any complications. In the placenta, fetal trophoblast cells come into contact with maternal immune cells. Inaccurate or inadequate adaptations of the maternal immune system could lead to problems with the functioning of the placenta. Macrophages are important for tissue homeostasis, cleanup, and the repair of damaged tissues. This is crucial for a rapidly developing organ such as the placenta. The consensus on macrophages at the maternal-fetal interface in pregnancy is that a major proportion have an anti-inflammatory, M2-like phenotype, that expresses scavenger receptors and is involved in tissue remodeling and the dampening of the immune reactions. Recent multidimensional analyses have contributed to a more detailed outlook on macrophages. The new view is that this lineage represents a highly diverse phenotype and is more prevalent than previously thought. Spatial-temporal in situ analyses during gestation have identified unique interactions of macrophages both with trophoblasts and with T cells at different trimesters of pregnancy. Here, we elaborate on the role of macrophages during early human pregnancy and at later gestation. Their possible effect is reviewed in the context of HLA incompatibility between mother and fetus, first in naturally conceived pregnancies, but foremost in pregnancies after oocyte donation. The potential functional consequences of macrophages for pregnancy-related immune reactions and the outcome in patients with recurrent pregnancy loss are also discussed.
Collapse
Affiliation(s)
- Juliette Krop
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Xuezi Tian
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Obstetrics and Gynaecology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marie-Louise van der Hoorn
- Department of Obstetrics and Gynaecology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
41
|
Chromogranin A: An Endocrine Factor of Pregnancy. Int J Mol Sci 2023; 24:ijms24054986. [PMID: 36902417 PMCID: PMC10002927 DOI: 10.3390/ijms24054986] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/26/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Pregnancy is a state of physiological and hormonal changes. One of the endocrine factors involved in these processes is chromogranin A, an acidic protein produced, among others, by the placenta. Although it has been previously linked to pregnancy, no existing articles have ever managed to clarify the role of this protein regarding this subject. Therefore, the aim of the present study is to gather knowledge of chromogranin A's function with reference to gestation and parturition, clarify elusive information, and, most importantly, to formulate hypotheses for the future studies to verify.
Collapse
|
42
|
Yu S, Huang C, Lian R, Diao L, Zhang X, Cai S, Wei H, Chen C, Li Y, Zeng Y. Establishment of reference intervals of endometrial immune cells during the mid-luteal phase. J Reprod Immunol 2023; 156:103822. [PMID: 36758471 DOI: 10.1016/j.jri.2023.103822] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/12/2023] [Accepted: 01/28/2023] [Indexed: 01/30/2023]
Abstract
This study aimed to develop reference intervals (RIs) of endometrial immune cells in control infertile women during the mid-luteal phase, and compare with the proportion of endometrial immune cells in recurrent reproductive failure (RRF) patients. Endometrial tissue sections were obtained from 113 fertile women and 79 patients with RRF, including 40 patients who had suffered recurrent miscarriage (RM) and 39 patients with repeated implantation failure (RIF) during the mid-luteal phase of the menstrual cycle. Immunohistochemical staining and quantitative analysis of CD56+, Foxp3+, CD163+, CD1a+ and CD8+ cells were performed in endometriums. RIs of endometrial immune cells in control infertile women were as follows: CD56+ uterine natural killer cells (uNK) cells, 1.785-8.712%, forkhead box P3 (Foxp3)+ Tregs, 0.041-0.154%, CD163+ M2 macrophages, 0.298-1.492%, CD1a+ dendritic cells (DCs), 0.006-0.081% and CD8+ T cells, 0.674-2.504%. Compared with control infertile women, the percentage of endometrial CD56+ uNK cells, CD163+ M2 macrophages, CD1a+ DCs and CD8+ T cells were significantly increased in patients with RRF. Moreover, Foxp3+ Tregs levels were decreased in patients with RRF, and were statistically significant only in patients with RM. In conclusion, the RIs of endometrial immune cells were established in control infertile women during the mid-luteal phase, and a disordered endometrial immune microenvironment was observed in patients with RRF. The RIs of endometrial immune cells may be of important clinical significance for the treatment of RRF.
Collapse
Affiliation(s)
- Shuyi Yu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Shenzhen Jinxin Medical Technology Innovation Center, Co., Ltd., Shenzhen, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation
| | - Chunyu Huang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Shenzhen Jinxin Medical Technology Innovation Center, Co., Ltd., Shenzhen, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation
| | - Ruochun Lian
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Shenzhen Jinxin Medical Technology Innovation Center, Co., Ltd., Shenzhen, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation
| | - Xueling Zhang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Shenzhen Jinxin Medical Technology Innovation Center, Co., Ltd., Shenzhen, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation
| | - Songchen Cai
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Shenzhen Jinxin Medical Technology Innovation Center, Co., Ltd., Shenzhen, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation
| | - Hongxia Wei
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Shenzhen Jinxin Medical Technology Innovation Center, Co., Ltd., Shenzhen, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation
| | - Cong Chen
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation
| | - Yuye Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Shenzhen Jinxin Medical Technology Innovation Center, Co., Ltd., Shenzhen, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation.
| | - Yong Zeng
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen 518045, Guangdong, China; Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-implantation.
| |
Collapse
|
43
|
Herrock O, Deer E, LaMarca B. Setting a stage: Inflammation during preeclampsia and postpartum. Front Physiol 2023; 14:1130116. [PMID: 36909242 PMCID: PMC9995795 DOI: 10.3389/fphys.2023.1130116] [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: 12/22/2022] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Preeclampsia (PE) is a leading cause of maternal and fetal mortality worldwide. The immune system plays a critical role in normal pregnancy progression; however, inappropriate inflammatory responses have been consistently linked with PE pathophysiology. This inflammatory phenotype consists of activation of the innate immune system, adaptive immune system, and increased inflammatory mediators in circulation. Moreover, recent studies have shown that the inflammatory profile seen in PE persists into the postpartum period. This manuscript aims to highlight recent advances in research relating to inflammation in PE as well as the inflammation that persists postpartum in women after a PE pregnancy. With the advent of the COVID-19 pandemic, there has been an increase in obstetric disorders associated with COVID-19 infection during pregnancy. This manuscript also aims to shed light on the relationship between COVID-19 infection during pregnancy and the increased incidence of PE in these women.
Collapse
Affiliation(s)
- Owen Herrock
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Evangeline Deer
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
| | - Babbette LaMarca
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Obstetrics and Gynecology, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
44
|
Chen Y, Xiao L, Xu J, Wang J, Yu Z, Zhao K, Zhang H, Cheng S, Sharma S, Liao A, Liu C. Recent insight into autophagy and immunity at the maternal-fetal interface. J Reprod Immunol 2023; 155:103781. [PMID: 36463798 DOI: 10.1016/j.jri.2022.103781] [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: 08/20/2022] [Revised: 11/01/2022] [Accepted: 11/27/2022] [Indexed: 11/30/2022]
Abstract
Autophagy is a lysosomal degradation pathway that supports metabolic adaptation and energy cycling. It is essential for cell homeostasis, differentiation, development, and survival. Recent studies have shown that autophagy could influence immune responses by regulating immune cell functions. Reciprocally, immune cells strongly influence autophagy. Immune cells at the maternal-fetal interface are thought to play essential roles in pregnancy. Here, we review the induction of autophagy at the maternal-fetal interface and its role in decidualization and placental development. Additionally, we emphasize the role of autophagy in the immune microenvironment at the maternal-fetal interface, including innate immunity, adaptive immunity, and immune tolerance molecules. It also suggests new research directions and prospects.
Collapse
Affiliation(s)
- Yuanyao Chen
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Lin Xiao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Jia Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Jingming Wang
- Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Zhiquan Yu
- Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China
| | - Shibin Cheng
- Department of Pediatrics, Obstetrics and Gynecology and Pathology, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Surendra Sharma
- Department of Pediatrics, Obstetrics and Gynecology and Pathology, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Aihua Liao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430030, PR China.
| |
Collapse
|
45
|
Genest G, Banjar S, Almasri W, Beauchamp C, Benoit J, Buckett W, Dzineku F, Gold P, Dahan MH, Jamal W, Jacques Kadoch I, Kadour-Peero E, Lapensée L, Miron P, Shaulov T, Sylvestre C, Tulandi T, Mazer BD, Laskin CA, Mahutte N. Immunomodulation for unexplained recurrent implantation failure: where are we now? Reproduction 2023; 165:R39-R60. [PMID: 36322478 DOI: 10.1530/rep-22-0150] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 11/02/2022] [Indexed: 11/05/2022]
Abstract
In brief Immune dysfunction may contribute to or cause recurrent implantation failure. This article summarizes normal and pathologic immune responses at implantation and critically appraises currently used immunomodulatory therapies. Abstract Recurrent implantation failure (RIF) may be defined as the absence of pregnancy despite the transfer of ≥3 good-quality blastocysts and is unexplained in up to 50% of cases. There are currently no effective treatments for patients with unexplained RIF. Since the maternal immune system is intricately involved in mediating endometrial receptivity and embryo implantation, both insufficient and excessive endometrial inflammatory responses during the window of implantation are proposed to lead to implantation failure. Recent strategies to improve conception rates in RIF patients have focused on modulating maternal immune responses at implantation, through either promoting or suppressing inflammation. Unfortunately, there are no validated, readily available diagnostic tests to confirm immune-mediated RIF. As such, immune therapies are often started empirically without robust evidence as to their efficacy. Like other chronic diseases, patient selection for immunomodulatory therapy is crucial, and personalized medicine for RIF patients is emerging. As the literature on the subject is heterogenous and rapidly evolving, we aim to summarize the potential efficacy, mechanisms of actions and side effects of select therapies for the practicing clinician.
Collapse
Affiliation(s)
- Geneviève Genest
- Department of Allergy and Immunology, McGill University, Montreal Quebec, Canada
| | - Shorooq Banjar
- Department of Allergy and Immunology, McGill University, Montreal Quebec, Canada
| | - Walaa Almasri
- Department of Allergy and Immunology, McGill University, Montreal Quebec, Canada
| | - Coralie Beauchamp
- Department of Gynaecology, University of Montreal, Montreal, Quebec, Canada
| | - Joanne Benoit
- Department of Gynaecology, University of Montreal, Montreal, Quebec, Canada
| | - William Buckett
- McGill University Health Centre Reproductive Centre, Montreal, Quebec, Canada
| | | | - Phil Gold
- Department of Allergy and Immunology, McGill University, Montreal Quebec, Canada
| | - Michael H Dahan
- Department of Obstetrics and Gynecology, McGill University, McGill University Health Centre, Montreal, Quebec, Canada
| | - Wael Jamal
- Department of Gynaecology, University of Montreal, Montreal, Quebec, Canada
| | | | - Einav Kadour-Peero
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, McGill University, Montréal, Quebec, Canada
| | - Louise Lapensée
- Department of Gynaecology, University of Montreal, Montreal, Quebec, Canada
| | - Pierre Miron
- Fertilys Reproductive Center, Laval, Quebec, Canada
| | - Talya Shaulov
- Department of Obstetrics and Gynecology, McGill University, McGill University Health Centre, Montreal, Quebec, Canada
| | - Camille Sylvestre
- Division of Reproductive Endocrinology and Infertility, University of Montreal, Montreal, Quebec, Canada
| | - Togas Tulandi
- Department of Obstetrics and Gynecology, McGill University, McGill University Health Centre, Montreal, Quebec, Canada
| | - Bruce D Mazer
- Department of Pediatrics, McGill University, Division of Allergy Immunology and Clinical Dermatology, Montreal Children's Hospital, McGill University, Montréal, Quebec, Canada
| | - Carl A Laskin
- Deptartments of Medicine and Obstetrics & Gynecology University of Toronto, Toronto, Ontario, Canada
| | - Neal Mahutte
- The Montreal Fertility Centre, Montreal, Quebec, Canada
| |
Collapse
|
46
|
Chen Q, Shan D, Xie Y, Luo X, Wu Y, Chen Q, Dong R, Hu Y. Single cell RNA sequencing research in maternal fetal interface. Front Cell Dev Biol 2023; 10:1079961. [PMID: 36704195 PMCID: PMC9871254 DOI: 10.3389/fcell.2022.1079961] [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: 10/25/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023] Open
Abstract
The maternal-fetal interface is an essential environment for embryonic growth and development, and a successful pregnancy depends on the dynamic balance of the microenvironment at the maternal-fetal interface. Single-cell sequencing, which unlike bulk sequencing that provides averaged data, is a robust method for interpreting the cellular and molecular landscape at single-cell resolution. With the support of single-cell sequencing, the issue of maternal-fetal interface heterogeneity during pregnancy has been more deeply elaborated and understood, which is important for a deeper understanding of physiological and pathological pregnancy. In this paper, we analyze the recent studies of single-cell transcriptomics in the maternal-fetal interface, and provide new directions for understanding and treating various pathological pregnancies.
Collapse
Affiliation(s)
- Qian Chen
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China,*Correspondence: Qian Chen, ; Yayi Hu,
| | - Dan Shan
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yupei Xie
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Xingrong Luo
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yuxia Wu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Qiuhe Chen
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ruihong Dong
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yayi Hu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China,Qingbaijiang Maternal and Child Health Hospital, Chengdu, China,*Correspondence: Qian Chen, ; Yayi Hu,
| |
Collapse
|
47
|
Sagae Y, Horie A, Yanai A, Ohara T, Nakakita B, Kitawaki Y, Okunomiya A, Tani H, Yamaguchi K, Hamanishi J, Lydon JP, Daikoku T, Watanabe H, Mandai M. Versican provides the provisional matrix for uterine spiral artery dilation and fetal growth. Matrix Biol 2023; 115:16-31. [PMID: 36423736 DOI: 10.1016/j.matbio.2022.11.004] [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: 08/02/2022] [Revised: 11/17/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022]
Abstract
The extracellular matrix (ECM) in the endometrium plays a crucial role in mammalian pregnancy. We have shown that versican secreted from the endometrial epithelium promotes embryo implantation. Versican is a proteoglycan, a major player in the provisional matrix, and versikine, its N-terminal fragment cleaved by ADAMTS proteinases, serves as a bioactive molecule. Here, since versican expression in the placenta was dynamically altered in humans and mice, we investigated the role of versican in pregnancy using uterine-specific Vcan deletion mice (uKO mice) and ADAMTS-resistant versican expressing mice (V1R mice). uKO mice exhibited insufficient spiral artery dilation, followed by fetal growth restriction and maternal hypertension. Further analysis revealed impaired proliferation of tissue-resident natural killer cells required for spiral artery dilation. V1R mice showed the same results as the control, eliminating the involvement of versikine. Our results provide a new concept that versican, one factor of ECM, contributes to placentation and following fetal growth.
Collapse
Affiliation(s)
- Yusuke Sagae
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihito Horie
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Akihiro Yanai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsutomu Ohara
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Baku Nakakita
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshimi Kitawaki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Asuka Okunomiya
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hirohiko Tani
- Department of Gynecology and Obstetrics, Shizuoka General Hospital, Shizuoka, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - John P Lydon
- Department of Pathology and Immunology, Center for Drug Discovery, Center for Reproductive Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Japan
| | - Hideto Watanabe
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
48
|
Covarrubias A, Aguilera-Olguín M, Carrasco-Wong I, Pardo F, Díaz-Astudillo P, Martín SS. Feto-placental Unit: From Development to Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:1-29. [PMID: 37466767 DOI: 10.1007/978-3-031-32554-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
The placenta is an intriguing organ that allows us to survive intrauterine life. This essential organ connects both mother and fetus and plays a crucial role in maternal and fetal well-being. This chapter presents an overview of the morphological and functional aspects of human placental development. First, we describe early human placental development and the characterization of the cell types found in the human placenta. Second, the human placenta from the second trimester to the term of gestation is reviewed, focusing on the morphology and specific pathologies that affect the placenta. Finally, we focus on the placenta's primary functions, such as oxygen and nutrient transport, and their importance for placental development.
Collapse
Affiliation(s)
- Ambart Covarrubias
- Health Sciences Faculty, Universidad San Sebastián, Concepción, Chile
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Macarena Aguilera-Olguín
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile
- Cellular Signalling and Differentiation Laboratory (CSDL), Medicine and Science Faculty, Universidad San Sebastián, Santiago, Chile
| | - Ivo Carrasco-Wong
- Cellular Signalling and Differentiation Laboratory (CSDL), School of Medical Technology, Medicine and Science Faculty, Universidad San Sebastián, Santiago, Chile
| | - Fabián Pardo
- Metabolic Diseases Research Laboratory, Interdisciplinary Centre of Territorial Health Research (CIISTe), Biomedical Research Center (CIB), San Felipe Campus, School of Medicine, Faculty of Medicine, Universidad de Valparaíso, San Felipe, Chile
| | - Pamela Díaz-Astudillo
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile
| | - Sebastián San Martín
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile.
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile.
| |
Collapse
|
49
|
Wei X, Yang X. The central role of natural killer cells in preeclampsia. Front Immunol 2023; 14:1009867. [PMID: 36865565 PMCID: PMC9972679 DOI: 10.3389/fimmu.2023.1009867] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Preeclampsia (PE) is a disease that is unique to pregnancy and affects multiple systems. It can lead to maternal and perinatal mortality. The precise etiology of PE is unclear. Patients with PE may have systemic or local immune abnormalities. A group of researchers has proposed that the immune communication between the fetus and mother is primarily moderated by natural killer (NK) cells as opposed to T cells, since NK cells are the most abundant immune cells in the uterus. This review examines the immunological roles of NK cells in the pathogenesis of PE. Our aim is to provide obstetricians with a comprehensive and updated research progress report on NK cells in PE patients. It has been reported that decidual NK (dNK) cells contribute to the process of uterine spiral artery remodeling and can modulate trophoblast invasion. Additionally, dNK cells can stimulate fetal growth and regulate delivery. It appears that the count or proportion of circulating NK cells is elevated in patients with or at risk for PE. Changes in the number or function of dNK cells may be the cause of PE. The Th1/Th2 equilibrium in PE has gradually shifted to an NK1/NK2 equilibrium based on cytokine production. An improper combination of killer cell immunoglobulin-like receptor (KIR) and human leukocyte antigen (HLA)-C may lead to insufficient activation of dNK cells, thereby causing PE. In the etiology of PE, NK cells appear to exert a central effect in both peripheral blood and the maternal-fetal interface. To maintain immune equilibrium both locally and systemically, it is necessary to take therapeutic measures directed at NK cells.
Collapse
Affiliation(s)
- Xiaoqi Wei
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| | - Xiuhua Yang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Gyselaers W. Origins of abnormal placentation: why maternal veins must not be forgotten. Am J Obstet Gynecol 2022:S0002-9378(22)02292-X. [PMID: 36539026 DOI: 10.1016/j.ajog.2022.12.014] [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: 09/17/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
The importance of uterine microvascular adaptations during placentation in pregnancy has been well established for decades. Inadequate dilatation of spiral arteries is associated with gestational complications, such as preeclampsia and/or intrauterine growth restriction. More recently, it has become clear that trophoblast cells invade and adapt decidual veins and lymphatic vessels 1 month before spiral arteries become patent and before intervillous space perfusion starts. Normal intervillous space hemodynamics is characterized by high volume flow at low velocity and pressure in the interseptal compartments surrounding the chorionic villi, hereby facilitating efficient maternal-fetal exchange. In case of shallow decidual vein dilatation, intervillous arterial supply exceeds venous drainage. This will cause congestion in the interseptal compartments with subsequently reduced perfusion and increased pressure. An efficient mechanism to counteract venous congestion and safeguard the viability of the conceptus is by reducing arterial inflow via shallow dilatation of the spiral arteries. This review made the case for intervillous space congestion as an unexplored trigger for inadequate spiral artery dilatation during the placentation process, eventually leading to abnormal systemic circulatory dysfunctions. An abnormal maternal venous function can result from an abnormal maternal immune response to paternal antigens with an imbalanced release of vasoactive mediators or can exist before conception. To get the full picture of abnormal placentation, maternal veins must not be forgotten.
Collapse
Affiliation(s)
- Wilfried Gyselaers
- Department of Obstetrics and Gynaecology, Ziekenhuis Oost Limburg, Genk, Belgium; Faculty Medicine and Life Sciences, Department of Physiology, Hasselt University, Diepenbeek, Belgium.
| |
Collapse
|