1
|
van Vliet MM, Boers RG, Boers JB, Schäffers OJM, van der Meeren LE, Steegers‐Theunissen RPM, Gribnau J, Schoenmakers S. Genome-wide methylation profiling of maternal cell-free DNA using methylated DNA sequencing (MeD-seq) indicates a placental and immune-cell signature. Eur J Clin Invest 2025; 55:e14363. [PMID: 39589853 PMCID: PMC11810559 DOI: 10.1111/eci.14363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Placental-originated cell-free DNA (cfDNA) provides unique opportunities to study (epi)genetic placental programming remotely, but studies investigating the cfDNA methylome are scarce and usually technologically challenging. Methylated DNA sequencing (MeD-seq) is well compatible with low cfDNA concentrations and has a high genome-wide coverage. We therefore aim to investigate the feasibility of genome-wide methylation profiling of first trimester maternal cfDNA using MeD-seq, by identifying placental-specific methylation marks in cfDNA. METHODS We collected cfDNA from nonpregnant controls (female n = 6, male n = 12) and pregnant women (n = 10), first trimester placentas (n = 10), and paired preconceptional and first trimester buffy coats (total n = 20). Differentially methylated regions (DMRs) were identified between pregnant and nonpregnant women. We investigated placental-specific markers in maternal cfDNA, including RASSF1 promoter and Y-chromosomal methylation, and studied overlap with placental and buffy coat DNA methylation. RESULTS We identified 436 DMRs between cfDNA from pregnant and nonpregnant women, which were validated using male cfDNA. RASSF1 promoter methylation was higher in maternal cfDNA (fold change 2.87, unpaired t-test p < .0001). Differential methylation of Y-chromosomal sequences could determine fetal sex. DMRs in maternal cfDNA showed large overlap with DNA methylation of these regions in placentas and buffy coats. Sixteen DMRs in maternal cfDNA were specifically found only in placentas. These novel potential placental-specific DMRs were more prominent than RASSF1. CONCLUSIONS MeD-seq can detect (novel) genome-wide placental DNA methylation marks and determine fetal sex in maternal cfDNA. Our results indicate a placental and immune-cell contribution to the pregnancy-specific cfDNA methylation signature. This study supports future research into maternal cfDNA methylation.
Collapse
Affiliation(s)
- Marjolein M. van Vliet
- Department of Obstetrics and GynaecologyErasmus MCRotterdamThe Netherlands
- Department of Developmental BiologyErasmus MCRotterdamThe Netherlands
| | - Ruben G. Boers
- Department of Developmental BiologyErasmus MCRotterdamThe Netherlands
| | - Joachim B. Boers
- Department of Developmental BiologyErasmus MCRotterdamThe Netherlands
| | - Olivier J. M. Schäffers
- Department of Obstetrics and GynaecologyErasmus MCRotterdamThe Netherlands
- Department of Developmental BiologyErasmus MCRotterdamThe Netherlands
| | - Lotte E. van der Meeren
- Department of PathologyErasmus Medical Centre RotterdamRotterdamThe Netherlands
- Department of PathologyLeiden University Medical CenterLeidenThe Netherlands
| | | | - Joost Gribnau
- Department of Developmental BiologyErasmus MCRotterdamThe Netherlands
| | - Sam Schoenmakers
- Department of Obstetrics and GynaecologyErasmus MCRotterdamThe Netherlands
| |
Collapse
|
2
|
Feng H, Tian C, Jiang W, Sun Z, Li Y, Han B, Chen L, Wang D, Xiang H, Zhu J, Song W, Li J, Cai Y, Wang S, Li Y. Hydrogen sulfide sustains mitochondria functions via targeting mitochondria fission regulator 1 like protein to restore human cytotrophoblast invasion and migration. Int J Biol Macromol 2025; 299:140240. [PMID: 39854860 DOI: 10.1016/j.ijbiomac.2025.140240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Hydrogen sulfide (H2S) is bioactive in mammals. Reduced H2S was observe in pregnancy complications, pre-eclampsia (PE). Our previous data demonstrated that low dose of H2S enhanced cytotrophoblast (CTB) invasion and migration via mitochondria dynamics without knowing the mechanisms. This study was designed to explore the functional regulation of CTB by mitochondrial fission regulator 1 like (MTFR1L) and the mechanisms. By studying human placenta samples and HTR-8/SVneo cell line, MTFR1L was found expressed in CTB. While MTFR1L expression was lower in PE placenta and CTB comparing with Normal pregnancy. Knockdown of MTFR1L decreased CTB invasion and migration, as well as the ATP production, while increased the mitochondria fragmentation, ROS production and mitochondria membrane potential indicating MTFR1L was key regulator of mitochondria. The posttranslational modulation analysis showed enhanced persulfidation of MTFR1L on cystine 222 and 230 by H2S. Mutations of MTFR1LC222/C230 suppressed ATP production, CTB invasion, migration, and increased mitochondria fragmentation, ROS production and mitochondria membrane potential. The present study showed the functional MTFR1L received endogenous CBS/H2S regulation. MTFR1LC222/230 persulfidation by H2S maintained mitochondria morphology and functions thus restored CTB invasion and migration. These findings established a new regulatory pathway for CTB invasion and migration, and provided new targets for PE treatment.
Collapse
Affiliation(s)
- Hao Feng
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Chunlei Tian
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Wenshan Jiang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250012, China
| | - Zongxin Sun
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Yikun Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Baoshi Han
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Lumei Chen
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Dawei Wang
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Hongjie Xiang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Jianchun Zhu
- Department of Pathology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Wengang Song
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yunlu Cai
- Laboratory of Metabolism and Gastrointestinal Tumor, The First Affiliated Hospital of Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Shuanglian Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250012, China.
| | - Yan Li
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Translational Medical Research Centre, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China.
| |
Collapse
|
3
|
Gehrs S, Jakab M, Gutjahr E, Gu Z, Weichenhan D, Mallm JP, Mogler C, Schlesner M, Plass C, Schlereth K, Augustin HG. The spatial zonation of the murine placental vasculature is specified by epigenetic mechanisms. Dev Cell 2025:S1534-5807(24)00776-7. [PMID: 39814015 DOI: 10.1016/j.devcel.2024.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 08/08/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
The labyrinthian fetoplacental capillary network is vital for proper nourishment of the developing embryo. Dysfunction of the maternal-fetal circulation is a primary cause of placental insufficiency. Here, we show that the spatial zonation of the murine placental labyrinth vasculature is controlled by flow-regulated epigenetic mechanisms. Spatiotemporal transcriptomic profiling identified a gradual change in the expression of epigenetic enzymes, including the de novo DNA methyltransferase 3a (DNMT3A). Loss of Dnmt3a resulted in DNA hypomethylation and perturbation of zonated placental gene expression. The resulting global DNA hypomethylation impaired the angiogenic capacity of endothelial cells. Global or endothelium-predominant deletion of Dnmt3a resulted in impaired placental vascularization and fetal growth retardation (FGR). Human placental endothelial gene expression profiling associated preeclampsia with reduced DNMT3A expression. Collectively, our study identified DMNT3A as critical methylome-regulator of placental endothelial gene expression and function with clinical implications for placental dysfunction, as it occurs during preeclampsia or FGR.
Collapse
Affiliation(s)
- Stephanie Gehrs
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany.
| | - Moritz Jakab
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ewgenija Gutjahr
- Institute of Pathology, University Clinic Heidelberg, 69120 Heidelberg, Germany
| | - Zuguang Gu
- Computational Oncology Group, Molecular Precision Oncology Program, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Dieter Weichenhan
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jan-Philipp Mallm
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, 69120 Heidelberg, Germany
| | - Carolin Mogler
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, 80333 Munich, Germany
| | - Matthias Schlesner
- Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, 86159 Augsburg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Katharina Schlereth
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| |
Collapse
|
4
|
Admati I, Skarbianskis N, Hochgerner H, Ophir O, Yagel S, Solt I, Zeisel A. Single-nuclei RNA-sequencing fails to detect molecular dysregulation in the preeclamptic placenta. Placenta 2025; 159:170-179. [PMID: 39733647 DOI: 10.1016/j.placenta.2024.12.011] [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: 07/22/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/31/2024]
Abstract
INTRODUCTION Single-cell RNA-seq (scRNA-seq) revolutionized our understanding of tissue complexity in health and disease and revealed massive transcriptional dysregulation across placental cell classes in early-onset, but not late-onset preeclampsia (PE). However, the multinucleated syncytium is largely inaccessible to cell dissociation. Nuclei isolation and single-nuclei RNA-seq may be preferable in the placenta; not least considering compatibility with long-term tissue storage. Yet, nuclei contain a subsample of the cells' transcriptional profile. Mature transcripts critical to cellular function and disease may be missed. METHODS We analyzed placenta from pregnancies using single-cell and single-nuclei RNA-seq. The datasets comprise 45,836 cells and 27,078 nuclei, from 10 to 7 early-onset preeclampsia (EPE) cases and 3 and 2 early idiopathic controls (ECT), respectively. We compared the methods' sensitivities, cell type detection, differential gene expression in PE, and performed histological validations. RESULTS Mature syncytiotrophoblast were sampled ∼50x more efficiently after nuclei extraction. Yet, scRNA-seq was more sensitive in detection of genes, molecules and mature transcripts. In snRNA-seq, nuclei of all placental cell classes suffered ambient trophoblast contamination. Transcripts from extravillous trophoblast, stroma, vasculature and immune cells were profiled less comprehensively by single-nuclei RNA-seq (snRNA-seq), restricting cell-type detection. In EPE, we found dysregulation of angiogenic actors FLT1/PGF both in prefused syncytiotrophoblast after cell extraction, and mature syncytiotrophoblast after nuclei isolation. Disease-related stress and inflammation were undetected from nuclei. DISCUSSION scRNA-seq has important advantages over snRNA-seq for comprehensive transcriptomics studies of the placenta, especially to understand cell-type resolved dysregulation in pathologies. Yet, to address the dilemma of an underrepresented syncytium, studies benefit from complementary nuclei extraction.
Collapse
Affiliation(s)
- Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Niv Skarbianskis
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Hannah Hochgerner
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Osnat Ophir
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Simcha Yagel
- Division of Obstetrics and Gynecology Hadassah, Hebrew University Medical Centers, Jerusalem, Israel
| | - Ido Solt
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel.
| | - Amit Zeisel
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
5
|
Zhou Z, Yang X. An update review of the application of single-cell RNA sequencing in pregnancy-related diseases. Front Endocrinol (Lausanne) 2024; 15:1415173. [PMID: 39717096 PMCID: PMC11663665 DOI: 10.3389/fendo.2024.1415173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 11/22/2024] [Indexed: 12/25/2024] Open
Abstract
Reproductive success hinges on the presence of a robust and functional placenta. Examining the placenta provides insight about the progression of pregnancy and valuable information about the normal developmental trajectory of the fetus. The current limitations of using bulk RNA-sequencing (RNA-seq) analysis stem from the diverse composition of the placenta, hindering a comprehensive description of how distinct trophoblast cell expression patterns contribute to the establishment and sustenance of a successful pregnancy. At present, the transcriptional landscape of intricate tissues increasingly relies on single-cell RNA sequencing (scRNA-seq). A few investigations have utilized scRNA-seq technology to examine the codes governing transcriptome regulation in cells at the maternal-fetal interface. In this review, we explore the fundamental principles of scRNA-seq technology, offering the latest overview of human placental studies utilizing this method across various gestational weeks in both normal pregnancies and pregnancy-related diseases, including recurrent pregnancy loss (RPL), preeclampsia (PE), preterm birth, and gestational diabetes mellitus (GDM). Furthermore, we discuss the limitations and future perspectives of scRNA-seq technology within the realm of reproduction. It seems that scRNA-seq stands out as one of the crucial tools for studying the etiology of pregnancy complications. The future direction of scRNA-seq applications may involve devolving into functional biology, with a primary focus on understanding variations in transcriptional activity among highly specific cell populations. Our goal is to provide obstetricians with an updated understanding of scRNA-seq technology related to pregnancy complications, providing comprehensive understandings to aid in the diagnosis and treatment of these conditions, ultimately improving maternal and fetal prognosis.
Collapse
Affiliation(s)
| | - Xiuhua Yang
- Department of Obstetrics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
6
|
Garcia-Flores V, Liu Z, Romero R, Pique-Regi R, Xu Y, Miller D, Levenson D, Galaz J, Winters AD, Farias-Jofre M, Panzer JJ, Theis KR, Gomez-Lopez N. Homeostatic Macrophages Prevent Preterm Birth and Improve Neonatal Outcomes by Mitigating In Utero Sterile Inflammation in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1620-1634. [PMID: 39431882 PMCID: PMC11572957 DOI: 10.4049/jimmunol.2400467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024]
Abstract
Preterm birth (PTB), often preceded by preterm labor, is a major cause of neonatal morbidity and mortality worldwide. Most PTB cases involve intra-amniotic inflammation without detectable microorganisms, termed in utero sterile inflammation, for which there is no established treatment. In this study, we propose homeostatic macrophages to prevent PTB and adverse neonatal outcomes caused by in utero sterile inflammation. Single-cell atlases of the maternal-fetal interface revealed that homeostatic maternal macrophages are reduced with human labor. M2 macrophage treatment prevented PTB and reduced adverse neonatal outcomes in mice with in utero sterile inflammation. Specifically, M2 macrophages halted premature labor by suppressing inflammatory responses in the amniotic cavity, including inflammasome activation, and mitigated placental and offspring lung inflammation. Moreover, M2 macrophages boosted gut inflammation in neonates and improved their ability to fight systemic bacterial infections. Our findings show that M2 macrophages are a promising strategy to mitigate PTB and improve neonatal outcomes resulting from in utero sterile inflammation.
Collapse
Affiliation(s)
- Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Zhenjie Liu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| | - Roger Pique-Regi
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Dustyn Levenson
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Andrew D. Winters
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Jonathan J. Panzer
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Kevin R. Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
7
|
Yoshida N, Thomas JR, Appios A, Brember MP, Aye IL, Edgar JR, Firth AE, Chung BY, McGovern N, Stewart H. Human placental cells are resistant to SARS-CoV-2 infection and replication. Wellcome Open Res 2024; 9:209. [PMID: 39640372 PMCID: PMC11617822 DOI: 10.12688/wellcomeopenres.20514.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
Background Infection during pregnancy with SARS-CoV-2 can have a serious impact on both maternal and foetal health. Clinical studies have shown that SARS-CoV-2 transmission from the mother to the foetus typically does not occur. However, there is evidence that SARS-CoV-2 can infect the placenta in utero. Here we sought to quantify the permissiveness of placental cells to SARS-CoV-2 infection and to determine if they support viral release. Methods By using publicly available single-cell RNA sequencing (scRNAseq) data sets and confocal microscopy we compared ACE2 transcript and protein expression across human first trimester and term placental cells. We also used in vitro infection assays to quantify the infection rates of a range of placenta-derived cells. Finally, we quantified the viral egress from these cells. Results ACE2 transcripts are found in a range of placental cell types across gestation, including trophoblast. However, ACE2 protein expression does not significantly change across placental cell types from first trimester to term. We find that 0.5±0.15 % of term trophoblast cells can be infected with SARS-CoV-2 while primary placental fibroblasts and macrophages, and JEG-3, JAR and HUVEC cell lines are resistant to infection. Furthermore, primary trophoblast cells poorly support viral release while JEG-3 cells allow relatively high levels of viral release. Conclusions The low level of viral release by primary placental cells provides insight into how the virus is impaired from crossing the placenta to the foetus.
Collapse
Affiliation(s)
- Nagisa Yoshida
- Department of Pathology, University of Cambridge, Cambridge, England, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, England, UK
| | - Jake R. Thomas
- Department of Pathology, University of Cambridge, Cambridge, England, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, England, UK
| | - Anna Appios
- Department of Pathology, University of Cambridge, Cambridge, England, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, England, UK
| | - Matthew P. Brember
- Department of Pathology, University of Cambridge, Cambridge, England, UK
| | - Irving L.M.H. Aye
- Centre for Trophoblast Research, University of Cambridge, Cambridge, England, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, England, UK
| | - James R. Edgar
- Department of Pathology, University of Cambridge, Cambridge, England, UK
| | - Andrew E. Firth
- Department of Pathology, University of Cambridge, Cambridge, England, UK
| | - Betty Y.W. Chung
- Department of Pathology, University of Cambridge, Cambridge, England, UK
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge, England, UK
- Centre for Trophoblast Research, University of Cambridge, Cambridge, England, UK
| | - Hazel Stewart
- Department of Pathology, University of Cambridge, Cambridge, England, UK
| |
Collapse
|
8
|
Tang M, Xiong L, Cai J, Fan L, Huang C, Zhang S, Jin Y, Luo E, Xing S, Yang X. Single-cell and spatial transcriptomics: Discovery of human placental development and disease. FASEB Bioadv 2024; 6:503-518. [PMID: 39512838 PMCID: PMC11539029 DOI: 10.1096/fba.2024-00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024] Open
Abstract
The human placenta is a vital organ, encompassing many distinct cell types, that maintains the growth and development of the fetus and is essential for substance exchange, defense, synthesis, and immunity. Abnormalities in placental cells can lead to various pregnancy complications, but the mechanisms remain largely unclear. Single-cell and spatial transcriptomics technologies have been developed in recent years to demonstrate placental cell heterogeneity and spatial molecular localization. Here, we review and summarize the current literature, demonstrating these technologies and showing the heterogeneity of various placenta cells and cell-cell communication of normal human placenta, as well as placenta-related diseases, such as preeclampsia, gestational diabetes mellitus, advanced maternal age, recurrent pregnancy loss, and placenta accreta spectrum disorders. Meanwhile, current weaknesses and future direction were discussed.
Collapse
Affiliation(s)
- Mi Tang
- Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Liling Xiong
- Obstetrics department, Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Jianghui Cai
- Department of Pharmacy, Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Li Fan
- Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Cheng Huang
- Clinical laboratory, Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Shimao Zhang
- Obstetrics department, Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Ying Jin
- Obstetrics department, Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Er‐dan Luo
- Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - ShaSha Xing
- Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| | - Xiao Yang
- Obstetrics department, Chengdu Women's and Children's Central Hospital, School of MedicineUniversity of Electronic Science and Technology of ChinaChengduChina
| |
Collapse
|
9
|
Zhou F, Sun Y, Chen X, Hou W, Shen J, Lai W, Han K, Zheng Y. Differences in cell subsets between sun-exposed and unexposed skin: preliminary single-cell sequencing and biological analysis from a single case. Front Med (Lausanne) 2024; 11:1453940. [PMID: 39540047 PMCID: PMC11558528 DOI: 10.3389/fmed.2024.1453940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction The composition and subsets of skin cells continuously change in a dynamic manner. However, the specific microcosmic alterations of human photoaged skin, independent of chronologic aging, remain unclear and have been infrequently analyzed. This study aimed to evaluate the biological processes and mechanisms underlying cell-subgroup alterations in skin photoaging. Methods We utilized single-cell sequencing and biological analysis from a single case to investigate the effects of photoaging. Skin punch biopsies were taken from sun-exposed forearm skin and unexposed buttock skin from the same individual for comparative analysis. Results Our analysis identified 25 cell clusters and 12 skin cell types, revealing significant changes in unique gene expressions between the sun-exposed and unexposed skin samples. A comparison of cell numbers within each cluster revealed 9 dominant cell clusters in sun-exposed skin and 16 dominant cell clusters in unexposed skin. Enrichment analysis indicated that PD-L1 expression and the PD-1 checkpoint pathway were more prominent in sun-exposed skin, while MAPK, TNF-alpha, TGF-beta, and apoptosis pathways were more enriched in hair follicle cells of sun-exposed skin. Discussion This study reveals changes in cell components in photoaged skin from a single case and provides novel insights into cellular subpopulations and pathology during repeated UVA-induced skin damage. These findings enhance our understanding of the complex interplay between different cells in photoaged skin and offer potential targets for preventing human skin photoaging and UV-induced skin cancers.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Sun
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinling Chen
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenyi Hou
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Shen
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Lai
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kai Han
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Zheng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Liu K, Wu S, Cui Y, Tao X, Li Y, Xiao X. Trophoblast fusion in fetal growth restriction is inhibited by CTGF in a cell-cycle-dependent manner. J Mol Histol 2024; 55:895-908. [PMID: 39122896 DOI: 10.1007/s10735-024-10239-9] [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: 02/03/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024]
Abstract
Fetal growth restriction (FGR) is a relatively common complication of pregnancy, and insufficient syncytialization in the placenta may play an important role in the pathogenesis of FGR. However, the mechanism of impaired formation of the syncytiotrophoblast layer in FGR patients requires further exploration. In the present study, we demonstrated that the level of syncytialization was decreased in FGR patient placentas, while the expression of connective tissue growth factor (CTGF) was significantly upregulated. CTGF was found to inhibit trophoblast fusion via regulating cell cycle progress of BeWo cells. Furthermore, we found that CTGF negatively regulates cell cycle arrest in a p21-dependent manner as overexpression of p21 could rescue the impaired syncytialization induced by CTGF-overexpression. Besides, we also identified that CTGF inhibits the expression of p21 through ITGB4/PI3K/AKT signaling pathway. Our study provided a new insight for elucidating the pathogenic mechanism of FGR and a novel idea for the clinical therapy of FGR.
Collapse
Affiliation(s)
- Ketong Liu
- Obstetrics and Gynecology Hospital of Fudan University, Fangxie Road 419, Shanghai, 200011, China
| | - Suwen Wu
- Obstetrics and Gynecology Hospital of Fudan University, Fangxie Road 419, Shanghai, 200011, China
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430050, China
| | - Yutong Cui
- Obstetrics and Gynecology Hospital of Fudan University, Fangxie Road 419, Shanghai, 200011, China
| | - Xiang Tao
- Obstetrics and Gynecology Hospital of Fudan University, Fangxie Road 419, Shanghai, 200011, China
| | - Yanhong Li
- Obstetrics and Gynecology Hospital of Fudan University, Fangxie Road 419, Shanghai, 200011, China.
| | - Xirong Xiao
- Obstetrics and Gynecology Hospital of Fudan University, Fangxie Road 419, Shanghai, 200011, China.
- Kashi Prefecture Second People's Hospital, Jiankang Road 1, Kashgar, 844000, China.
| |
Collapse
|
11
|
Cao C, Saxena R, Gray KJ. Placental Origins of Preeclampsia: Insights from Multi-Omic Studies. Int J Mol Sci 2024; 25:9343. [PMID: 39273292 PMCID: PMC11395466 DOI: 10.3390/ijms25179343] [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: 07/22/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Preeclampsia (PE) is a major cause of maternal and neonatal morbidity and mortality worldwide, with the placenta playing a central role in disease pathophysiology. This review synthesizes recent advancements in understanding the molecular mechanisms underlying PE, focusing on placental genes, proteins, and genetic variants identified through multi-omic approaches. Transcriptomic studies in bulk placental tissue have identified many dysregulated genes in the PE placenta, including the PE signature gene, Fms-like tyrosine kinase 1 (FLT1). Emerging single-cell level transcriptomic data have revealed key cell types and molecular signatures implicated in placental dysfunction and PE. However, the considerable variability among studies underscores the need for standardized methodologies and larger sample sizes to enhance the reproducibility of results. Proteomic profiling of PE placentas has identified numerous PE-associated proteins, offering insights into potential biomarkers and pathways implicated in PE pathogenesis. Despite significant progress, challenges such as inconsistencies in study findings and lack of validation persist. Recent fetal genome-wide association studies have identified multiple genetic loci associated with PE, with ongoing efforts to elucidate their impact on placental gene expression and function. Future directions include the integration of multi-omic data, validation of findings in diverse PE populations and clinical subtypes, and the development of analytical approaches and experimental models to study the complex interplay of placental and maternal factors in PE etiology. These insights hold promise for improving risk prediction, diagnosis, and management of PE, ultimately reducing its burden on maternal and neonatal health.
Collapse
Affiliation(s)
- Chang Cao
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Richa Saxena
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kathryn J. Gray
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
12
|
Van Buren E, Azzara D, Rangel-Moreno J, Garcia-Hernandez MDLL, Murphy SP, Cohen ED, Lewis E, Lin X, Park HR. Single-cell RNA sequencing reveals placental response under environmental stress. Nat Commun 2024; 15:6549. [PMID: 39095385 PMCID: PMC11297347 DOI: 10.1038/s41467-024-50914-9] [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: 08/16/2023] [Accepted: 07/25/2024] [Indexed: 08/04/2024] Open
Abstract
The placenta is crucial for fetal development, yet the impact of environmental stressors such as arsenic exposure remains poorly understood. We apply single-cell RNA sequencing to analyze the response of the mouse placenta to arsenic, revealing cell-type-specific gene expression, function, and pathological changes. Notably, the Prap1 gene, which encodes proline-rich acidic protein 1 (PRAP1), is significantly upregulated in 26 placental cell types including various trophoblast cells. Our study shows a female-biased increase in PRAP1 in response to arsenic and localizes it in the placenta. In vitro and ex vivo experiments confirm PRAP1 upregulation following arsenic treatment and demonstrate that recombinant PRAP1 protein reduces arsenic-induced cytotoxicity and downregulates cell cycle pathways in human trophoblast cells. Moreover, PRAP1 knockdown differentially affects cell cycle processes, proliferation, and cell death depending on the presence of arsenic. Our findings provide insights into the placental response to environmental stress, offering potential preventative and therapeutic approaches for environment-related adverse outcomes in mothers and children.
Collapse
Affiliation(s)
- Eric Van Buren
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - David Azzara
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Javier Rangel-Moreno
- Division of Allergy, Immunology and Rheumatology, Department of Medicine, University of Rochester, Rochester, NY, USA
| | | | - Shawn P Murphy
- Department of Obstetrics and Gynecology, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Ethan D Cohen
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Ethan Lewis
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
| | - Hae-Ryung Park
- Department of Environmental Medicine, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
13
|
Ackerman WE, Rigo MM, DaSilva-Arnold SC, Do C, Tariq M, Salas M, Castano A, Zamudio S, Tycko B, Illsley NP. Epigenetic changes regulating the epithelial-mesenchymal transition in human trophoblast differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601748. [PMID: 39005325 PMCID: PMC11244995 DOI: 10.1101/2024.07.02.601748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The phenotype of human placental extravillous trophoblast (EVT) at the end of pregnancy reflects both first trimester differentiation from villous cytotrophoblast (CTB) and later gestational changes, including loss of proliferative and invasive capacity. Invasion abnormalities are central to two major placental pathologies, preeclampsia and placenta accreta spectrum, so characterization of the corresponding normal processes is crucial. In this report, our gene expression analysis, using purified human CTB and EVT cells, highlights an epithelial-mesenchymal transition (EMT) mechanism underlying CTB-EVT differentiation and provides a trophoblast-specific EMT signature. In parallel, DNA methylation profiling shows that CTB cells, already hypomethylated relative to non-trophoblast cell lineages, show further genome-wide hypomethylation in the transition to EVT. However, a small subgroup of genes undergoes gains of methylation (GOM) in their regulatory regions or gene bodies, associated with differential mRNA expression (DE). Prominent in this GOM-DE group are genes involved in the EMT, including multiple canonical EMT markers and the EMT-linked transcription factor RUNX1, for which we demonstrate a functional role in modulating the migratory and invasive capacities of JEG3 trophoblast cells. This analysis of DE associated with locus-specific GOM, together with functional studies of an important GOM-DE gene, highlights epigenetically regulated genes and pathways acting in human EVT differentiation and invasion, with implications for obstetric disorders in which these processes are dysregulated.
Collapse
Affiliation(s)
- William E. Ackerman
- Department of Obstetrics and Gynecology and AI.Health4All Center for Health Equity Using Machine Learning and Artificial Intelligence, University of Illinois College of Medicine, Chicago, USA
| | - Mauricio M. Rigo
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ
| | - Sonia C. DaSilva-Arnold
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack NJ
| | - Catherine Do
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ
| | - Mariam Tariq
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ
| | - Martha Salas
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ
| | - Angelica Castano
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ
| | - Stacy Zamudio
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack NJ
| | - Benjamin Tycko
- Hackensack Meridian Health Center for Discovery and Innovation, Nutley, NJ
| | - Nicholas P. Illsley
- Department of Obstetrics and Gynecology, Hackensack University Medical Center, Hackensack NJ
| |
Collapse
|
14
|
Derisoud E, Jiang H, Zhao A, Chavatte-Palmer P, Deng Q. Revealing the molecular landscape of human placenta: a systematic review and meta-analysis of single-cell RNA sequencing studies. Hum Reprod Update 2024; 30:410-441. [PMID: 38478759 PMCID: PMC11215163 DOI: 10.1093/humupd/dmae006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 02/12/2024] [Indexed: 07/02/2024] Open
Abstract
BACKGROUND With increasing significance of developmental programming effects associated with placental dysfunction, more investigations are devoted to improving the characterization and understanding of placental signatures in health and disease. The placenta is a transitory but dynamic organ adapting to the shifting demands of fetal development and available resources of the maternal supply throughout pregnancy. Trophoblasts (cytotrophoblasts, syncytiotrophoblasts, and extravillous trophoblasts) are placental-specific cell types responsible for the main placental exchanges and adaptations. Transcriptomic studies with single-cell resolution have led to advances in understanding the placenta's role in health and disease. These studies, however, often show discrepancies in characterization of the different placental cell types. OBJECTIVE AND RATIONALE We aim to review the knowledge regarding placental structure and function gained from the use of single-cell RNA sequencing (scRNAseq), followed by comparing cell-type-specific genes, highlighting their similarities and differences. Moreover, we intend to identify consensus marker genes for the various trophoblast cell types across studies. Finally, we will discuss the contributions and potential applications of scRNAseq in studying pregnancy-related diseases. SEARCH METHODS We conducted a comprehensive systematic literature review to identify different cell types and their functions at the human maternal-fetal interface, focusing on all original scRNAseq studies on placentas published before March 2023 and published reviews (total of 28 studies identified) using PubMed search. Our approach involved curating cell types and subtypes that had previously been defined using scRNAseq and comparing the genes used as markers or identified as potential new markers. Next, we reanalyzed expression matrices from the six available scRNAseq raw datasets with cell annotations (four from first trimester and two at term), using Wilcoxon rank-sum tests to compare gene expression among studies and annotate trophoblast cell markers in both first trimester and term placentas. Furthermore, we integrated scRNAseq raw data available from 18 healthy first trimester and nine term placentas, and performed clustering and differential gene expression analysis. We further compared markers obtained with the analysis of annotated and raw datasets with the literature to obtain a common signature gene list for major placental cell types. OUTCOMES Variations in the sampling site, gestational age, fetal sex, and subsequent sequencing and analysis methods were observed between the studies. Although their proportions varied, the three trophoblast types were consistently identified across all scRNAseq studies, unlike other non-trophoblast cell types. Notably, no marker genes were shared by all studies for any of the investigated cell types. Moreover, most of the newly defined markers in one study were not observed in other studies. These discrepancies were confirmed by our analysis on trophoblast cell types, where hundreds of potential marker genes were identified in each study but with little overlap across studies. From 35 461 and 23 378 cells of high quality in the first trimester and term placentas, respectively, we obtained major placental cell types, including perivascular cells that previously had not been identified in the first trimester. Importantly, our meta-analysis provides marker genes for major placental cell types based on our extensive curation. WIDER IMPLICATIONS This review and meta-analysis emphasizes the need for establishing a consensus for annotating placental cell types from scRNAseq data. The marker genes identified here can be deployed for defining human placental cell types, thereby facilitating and improving the reproducibility of trophoblast cell annotation.
Collapse
Affiliation(s)
- Emilie Derisoud
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Hong Jiang
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Allan Zhao
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
| | - Pascale Chavatte-Palmer
- INRAE, BREED, Université Paris-Saclay, UVSQ, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d’Alfort, BREED, Maisons-Alfort, France
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska University Hospital, Solna, Stockholm, Sweden
| |
Collapse
|
15
|
Batorsky R, Ceasrine AM, Shook LL, Kislal S, Bordt EA, Devlin BA, Perlis RH, Slonim DK, Bilbo SD, Edlow AG. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. Cell Rep 2024; 43:114326. [PMID: 38848212 PMCID: PMC11808824 DOI: 10.1016/j.celrep.2024.114326] [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: 12/13/2023] [Revised: 04/25/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Maternal immune activation is associated with adverse offspring neurodevelopmental outcomes, many mediated by in utero microglial programming. As microglia remain inaccessible throughout development, identification of noninvasive biomarkers reflecting fetal brain microglial programming could permit screening and intervention. We used lineage tracing to demonstrate the shared ontogeny between fetal brain macrophages (microglia) and fetal placental macrophages (Hofbauer cells) in a mouse model of maternal diet-induced obesity, and single-cell RNA-seq to demonstrate shared transcriptional programs. Comparison with human datasets demonstrated conservation of placental resident macrophage signatures between mice and humans. Single-cell RNA-seq identified common alterations in fetal microglial and Hofbauer cell gene expression induced by maternal obesity, as well as sex differences in these alterations. We propose that Hofbauer cells, which are easily accessible at birth, provide insights into fetal brain microglial programs and may facilitate the early identification of offspring vulnerable to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rebecca Batorsky
- Data Intensive Studies Center, Tufts University, Medford, MA, USA
| | - Alexis M Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA
| | - Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin A Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Roy H Perlis
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA, USA
| | - Staci D Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA; Department of Neurobiology, Duke University, Durham, NC, USA; Lurie Center for Autism, Massachusetts General Hospital, Boston, MA, USA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
16
|
Yue T, Guo Y, Qi X, Zheng W, Zhang H, Wang B, Liu K, Zhou B, Zeng X, Ouzhuluobu, He Y, Su B. Sex-biased regulatory changes in the placenta of native highlanders contribute to adaptive fetal development. eLife 2024; 12:RP89004. [PMID: 38869160 PMCID: PMC11175615 DOI: 10.7554/elife.89004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
Compared with lowlander migrants, native Tibetans have a higher reproductive success at high altitude though the underlying mechanism remains unclear. Here, we compared the transcriptome and histology of full-term placentas between native Tibetans and Han migrants. We found that the placental trophoblast shows the largest expression divergence between Tibetans and Han, and Tibetans show decreased immune response and endoplasmic reticulum stress. Remarkably, we detected a sex-biased expression divergence, where the male-infant placentas show a greater between-population difference than the female-infant placentas. The umbilical cord plays a key role in the sex-biased expression divergence, which is associated with the higher birth weight of the male newborns of Tibetans. We also identified adaptive histological changes in the male-infant placentas of Tibetans, including larger umbilical artery wall and umbilical artery intima and media, and fewer syncytial knots. These findings provide valuable insights into the sex-biased adaptation of human populations, with significant implications for medical and genetic studies of human reproduction.
Collapse
Affiliation(s)
- Tian Yue
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Kunming College of Life Science, University of Chinese Academy of SciencesBeijingChina
| | - Yongbo Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Kunming College of Life Science, University of Chinese Academy of SciencesBeijingChina
| | - Xuebin Qi
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang HospitalKunmingChina
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
| | - Wangshan Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
| | - Hui Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyKunmingChina
| | - Bin Wang
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang HospitalKunmingChina
| | - Kai Liu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
| | - Bin Zhou
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Kunming College of Life Science, University of Chinese Academy of SciencesBeijingChina
| | - Xuerui Zeng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Kunming College of Life Science, University of Chinese Academy of SciencesBeijingChina
| | - Ouzhuluobu
- Fukang Obstetrics, Gynecology and Children Branch Hospital, Tibetan Fukang HospitalKunmingChina
| | - Yaoxi He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
| | - Bing Su
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of SciencesKunmingChina
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of SciencesKunmingChina
| |
Collapse
|
17
|
Barrozo ER, Racusin DA, Jochum MD, Garcia BT, Suter MA, Delbeccaro M, Shope C, Antony K, Aagaard KM. Discrete placental gene expression signatures accompany diabetic disease classifications during pregnancy. Am J Obstet Gynecol 2024:S0002-9378(24)00596-9. [PMID: 38763341 DOI: 10.1016/j.ajog.2024.05.014] [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: 12/28/2023] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Gestational diabetes mellitus affects up to 10% of pregnancies and is classified into subtypes gestational diabetes subtype A1 (GDMA1) (managed by lifestyle modifications) and gestational diabetes subtype A2 (GDMA2) (requiring medication). However, whether these subtypes are distinct clinical entities or more reflective of an extended spectrum of normal pregnancy endocrine physiology remains unclear. OBJECTIVE Integrated bulk RNA-sequencing (RNA-seq), single-cell RNA-sequencing (scRNA-seq), and spatial transcriptomics harbors the potential to reveal disease gene signatures in subsets of cells and tissue microenvironments. We aimed to combine these high-resolution technologies with rigorous classification of diabetes subtypes in pregnancy. We hypothesized that differences between preexisting type 2 and gestational diabetes subtypes would be associated with altered gene expression profiles in specific placental cell populations. STUDY DESIGN In a large case-cohort design, we compared validated cases of GDMA1, GDMA2, and type 2 diabetes mellitus (T2DM) to healthy controls by bulk RNA-seq (n=54). Quantitative analyses with reverse transcription and quantitative PCR of presumptive genes of significant interest were undertaken in an independent and nonoverlapping validation cohort of similarly well-characterized cases and controls (n=122). Additional integrated analyses of term placental single-cell, single-nuclei, and spatial transcriptomics data enabled us to determine the cellular subpopulations and niches that aligned with the GDMA1, GDMA2, and T2DM gene expression signatures at higher resolution and with greater confidence. RESULTS Dimensional reduction of the bulk RNA-seq data revealed that the most common source of placental gene expression variation was the diabetic disease subtype. Relative to controls, we found 2052 unique and significantly differentially expressed genes (-22 thresholds; q<0.05 Wald Test) among GDMA1 placental specimens, 267 among GDMA2, and 1520 among T2DM. Several candidate marker genes (chorionic somatomammotropin hormone 1 [CSH1], period circadian regulator 1 [PER1], phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta [PIK3CB], forkhead box O1 [FOXO1], epidermal growth factor receptor [EGFR], interleukin 2 receptor subunit beta [IL2RB], superoxide dismutase 3 [SOD3], dedicator of cytokinesis 5 [DOCK5], suppressor of glucose, and autophagy associated 1 [SOGA1]) were validated in an independent and nonoverlapping validation cohort (q<0.05 Tukey). Functional enrichment revealed the pathways and genes most impacted for each diabetes subtype, and the degree of proximal similarity to other subclassifications. Surprisingly, GDMA1 and T2DM placental signatures were more alike by virtue of increased expression of chromatin remodeling and epigenetic regulation genes, while albumin was the top marker for GDMA2 with increased expression of placental genes in the wound healing pathway. Assessment of these gene signatures in single-cell, single-nuclei, and spatial transcriptomics data revealed high specificity and variability by placental cell and microarchitecture types. For example, at the cellular and spatial (eg, microarchitectural) levels, distinguishing features were observed in extravillous trophoblasts (GDMA1) and macrophages (GDMA2). Lastly, we utilized these data to train and evaluate 4 machine learning models to estimate our confidence in predicting the control or diabetes status of placental transcriptome specimens with no available clinical metadata. CONCLUSION Consistent with the distinct association of perinatal outcome risk, placentae from GDMA1, GDMA2, and T2DM-affected pregnancies harbor unique gene signatures that can be further distinguished by altered placental cellular subtypes and microarchitectural niches.
Collapse
Affiliation(s)
- Enrico R Barrozo
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Diana A Racusin
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Michael D Jochum
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Brandon T Garcia
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX; Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, TX
| | - Melissa A Suter
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Melanie Delbeccaro
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Cynthia Shope
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Kathleen Antony
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX.
| |
Collapse
|
18
|
Shu C, Street K, Breton CV, Bastain TM, Wilson ML. A review of single-cell transcriptomics and epigenomics studies in maternal and child health. Epigenomics 2024; 16:775-793. [PMID: 38709139 PMCID: PMC11318716 DOI: 10.1080/17501911.2024.2343276] [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] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
Single-cell sequencing technologies enhance our understanding of cellular dynamics throughout pregnancy. We outlined the workflow of single-cell sequencing techniques and reviewed single-cell studies in maternal and child health. We conducted a literature review of single cell studies on maternal and child health using PubMed. We summarized the findings from 16 single-cell atlases of the human and mammalian placenta across gestational stages and 31 single-cell studies on maternal exposures and complications including infection, obesity, diet, gestational diabetes, pre-eclampsia, environmental exposure and preterm birth. Single-cell studies provides insights on novel cell types in placenta and cell type-specific marks associated with maternal exposures and complications.
Collapse
Affiliation(s)
- Chang Shu
- Center for Genetic Epidemiology, Division of Epidemiology & Genetics, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Kelly Street
- Division of Biostatistics, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Carrie V Breton
- Division of Environmental Health, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Theresa M Bastain
- Division of Environmental Health, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Melissa L Wilson
- Division of Disease Prevention, Policy, & Global Health, Department of Population & Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles,CA USA
| |
Collapse
|
19
|
Afshar Y, Yin O, Jeong A, Martinez G, Kim J, Ma F, Jang C, Tabatabaei S, You S, Tseng HR, Zhu Y, Krakow D. Placenta accreta spectrum disorder at single-cell resolution: a loss of boundary limits in the decidua and endothelium. Am J Obstet Gynecol 2024; 230:443.e1-443.e18. [PMID: 38296740 DOI: 10.1016/j.ajog.2023.10.001] [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: 06/06/2023] [Revised: 09/25/2023] [Accepted: 10/01/2023] [Indexed: 02/02/2024]
Abstract
BACKGROUND Placenta accreta spectrum disorders are associated with severe maternal morbidity and mortality. Placenta accreta spectrum disorders involve excessive adherence of the placenta preventing separation at birth. Traditionally, this condition has been attributed to excessive trophoblast invasion; however, an alternative view is a fundamental defect in decidual biology. OBJECTIVE This study aimed to gain insights into the understanding of placenta accreta spectrum disorder by using single-cell and spatially resolved transcriptomics to characterize cellular heterogeneity at the maternal-fetal interface in placenta accreta spectrum disorders. STUDY DESIGN To assess cellular heterogeneity and the function of cell types, single-cell RNA sequencing and spatially resolved transcriptomics were used. A total of 12 placentas were included, 6 placentas with placenta accreta spectrum disorder and 6 controls. For each placenta with placenta accreta spectrum disorder, multiple biopsies were taken at the following sites: placenta accreta spectrum adherent and nonadherent sites in the same placenta. Of note, 2 platforms were used to generate libraries: the 10× Chromium and NanoString GeoMX Digital Spatial Profiler for single-cell and spatially resolved transcriptomes, respectively. Differential gene expression analysis was performed using a suite of bioinformatic tools (Seurat and GeoMxTools R packages). Correction for multiple testing was performed using Clipper. In situ hybridization was performed with RNAscope, and immunohistochemistry was used to assess protein expression. RESULTS In creating a placenta accreta cell atlas, there were dramatic difference in the transcriptional profile by site of biopsy between placenta accreta spectrum and controls. Most of the differences were noted at the site of adherence; however, differences existed within the placenta between the adherent and nonadherent site of the same placenta in placenta accreta. Among all cell types, the endothelial-stromal populations exhibited the greatest difference in gene expression, driven by changes in collagen genes, namely collagen type III alpha 1 chain (COL3A1), growth factors, epidermal growth factor-like protein 6 (EGFL6), and hepatocyte growth factor (HGF), and angiogenesis-related genes, namely delta-like noncanonical Notch ligand 1 (DLK1) and platelet endothelial cell adhesion molecule-1 (PECAM1). Intraplacental tropism (adherent versus non-adherent sites in the same placenta) was driven by differences in endothelial-stromal cells with notable differences in bone morphogenic protein 5 (BMP5) and osteopontin (SPP1) in the adherent vs nonadherent site of placenta accreta spectrum. CONCLUSION Placenta accreta spectrum disorders were characterized at single-cell resolution to gain insight into the pathophysiology of the disease. An atlas of the placenta at single cell resolution in accreta allows for understanding in the biology of the intimate maternal and fetal interaction. The contributions of stromal and endothelial cells were demonstrated through alterations in the extracellular matrix, growth factors, and angiogenesis. Transcriptional and protein changes in the stroma of placenta accreta spectrum shift the etiologic explanation away from "invasive trophoblast" to "loss of boundary limits" in the decidua. Gene targets identified in this study may be used to refine diagnostic assays in early pregnancy, track disease progression over time, and inform therapeutic discoveries.
Collapse
Affiliation(s)
- Yalda Afshar
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA.
| | - Ophelia Yin
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA; Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, San Francisco, CA
| | - Anhyo Jeong
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Guadalupe Martinez
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Jina Kim
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Feiyang Ma
- Department of Molecular, Cell, and Developmental Biology, University of California, Los Angeles, Los Angeles, CA
| | - Christine Jang
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Sarah Tabatabaei
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| | - Sungyong You
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA; Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, Los Angeles, CA
| | - Yazhen Zhu
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, Los Angeles, CA; Department of Pathology, University of California, Los Angeles, Los Angeles, CA
| | - Deborah Krakow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA; Departments of Orthopedic Surgery and Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
20
|
Williams RP, Lesseur C, Cheng H, Li Q, Deyssenroth M, Molteno CD, Meintjes EM, Jacobson SW, Jacobson JL, Wainwright H, Hao K, Chen J, Carter RC. RNA-seq analysis reveals prenatal alcohol exposure is associated with placental inflammatory cells and gene expression. Gene 2024; 894:147951. [PMID: 37918548 PMCID: PMC11819563 DOI: 10.1016/j.gene.2023.147951] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/21/2023] [Accepted: 10/30/2023] [Indexed: 11/04/2023]
Abstract
BACKGROUND Fetal alcohol spectrum disorders (FASD) are the most common preventable cause of birth defects and neurodevelopmental disorders worldwide. The placenta is the crucial interface between mother and fetus. Prenatal alcohol exposure (PAE) has been shown to alter placental structure and expression of genes in bulk placental tissue samples, but prior studies have not examined effects on placental cell-type composition or taken cell-type into consideration in transcriptome analyses. METHODS We leveraged an existent placenta single-cell RNA-seq dataset to perform cell-type deconvolution of bulk placental RNA-seq data from 35 heavy drinking pregnant women and 33 controls in a prospective birth cohort in Cape Town, South Africa. We used bivariate analyses and multivariable adjusted linear regression models to assess the relation of PAE on inferred placental cell-type proportions. We also examined differential expression of inflammatory response genes and PAE, using multivariable adjusted linear models. RESULTS Deconvolution analyses showed heterogeneous placenta cell-type composition in which stromal (27 %), endothelial (26 %) and cytotrophoblasts (18 %) were the predominant cell-types. PAE around conception was associated with a higher proportion of Hofbauer cells (B = 0.51, p = 0.035) in linear models adjusted for maternal age, infant sex, and gestational age. Among the 652 inflammatory genes examined, 35 were differential expressed in alcohol exposed placentas (FDR p < 0.05). CONCLUSIONS Our findings suggest that heavy alcohol exposure during pregnancy can influence the proportion of fetal placental villi macrophages (Hofbauer cells) and increased expression of inflammatory genes. Future studies are needed to further characterize these effects and to assess the potential functional roles of placental inflammation in FASD.
Collapse
Affiliation(s)
- Randy P Williams
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Corina Lesseur
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Haoxiang Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qian Li
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maya Deyssenroth
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Christopher D Molteno
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Ernesta M Meintjes
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa
| | - Sandra W Jacobson
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa; Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Joseph L Jacobson
- Department of Psychiatry and Mental Health, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA
| | - Helen Wainwright
- Department of Pathology, National Health Laboratory Service, Cape Town, South Africa
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - R Colin Carter
- Department of Human Biology, University of Cape Town Faculty of Health Sciences, Cape Town, South Africa; Departments of Emergency Medicine and Pediatrics, Institute of Human Nutrition, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
21
|
Wang M, Liu Y, Sun R, Liu F, Li J, Yan L, Zhang J, Xie X, Li D, Wang Y, Li S, Zhu X, Li R, Lu F, Xiao Z, Wang H. Single-nucleus multi-omic profiling of human placental syncytiotrophoblasts identifies cellular trajectories during pregnancy. Nat Genet 2024; 56:294-305. [PMID: 38267607 PMCID: PMC10864176 DOI: 10.1038/s41588-023-01647-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: 07/26/2022] [Accepted: 12/11/2023] [Indexed: 01/26/2024]
Abstract
The human placenta has a vital role in ensuring a successful pregnancy. Despite the growing body of knowledge about its cellular compositions and functions, there has been limited research on the heterogeneity of the billions of nuclei within the syncytiotrophoblast (STB), a multinucleated entity primarily responsible for placental function. Here we conducted integrated single-nucleus RNA sequencing and single-nucleus ATAC sequencing analyses of human placentas from early and late pregnancy. Our findings demonstrate the dynamic heterogeneity and developmental trajectories of STB nuclei and their correspondence with human trophoblast stem cell (hTSC)-derived STB. Furthermore, we identified transcription factors associated with diverse STB nuclear lineages through their gene regulatory networks and experimentally confirmed their function in hTSC and trophoblast organoid-derived STBs. Together, our data provide insights into the heterogeneity of human STB and represent a valuable resource for interpreting associated pregnancy complications.
Collapse
Affiliation(s)
- Meijiao Wang
- The Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yawei Liu
- The Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
- Medical Center of Soochow University, Suzhou, China
- Suzhou Dushu Lake Hospital, Suzhou, China
| | - Run Sun
- The Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fenting Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China
| | - Jiaqian Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Long Yan
- The Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jixiang Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xinwei Xie
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Dongxu Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Yiming Wang
- The Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shiwen Li
- The Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xili Zhu
- The Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China.
| | - Falong Lu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Zhenyu Xiao
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- School of Life Science, Beijing Institute of Technology, Beijing, China.
| | - Hongmei Wang
- The Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
22
|
Li Q, Sharkey A, Sheridan M, Magistrati E, Arutyunyan A, Huhn O, Sancho-Serra C, Anderson H, McGovern N, Esposito L, Fernando R, Gardner L, Vento-Tormo R, Turco MY, Moffett A. Human uterine natural killer cells regulate differentiation of extravillous trophoblast early in pregnancy. Cell Stem Cell 2024; 31:181-195.e9. [PMID: 38237587 DOI: 10.1016/j.stem.2023.12.013] [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: 05/13/2023] [Revised: 10/19/2023] [Accepted: 12/20/2023] [Indexed: 02/04/2024]
Abstract
In humans, balanced invasion of trophoblast cells into the uterine mucosa, the decidua, is critical for successful pregnancy. Evidence suggests that this process is regulated by uterine natural killer (uNK) cells, but how they influence reproductive outcomes is unclear. Here, we used our trophoblast organoids and primary tissue samples to determine how uNK cells affect placentation. By locating potential interaction axes between trophoblast and uNK cells using single-cell transcriptomics and in vitro modeling of these interactions in organoids, we identify a uNK cell-derived cytokine signal that promotes trophoblast differentiation at the late stage of the invasive pathway. Moreover, it affects transcriptional programs involved in regulating blood flow, nutrients, and inflammatory and adaptive immune responses, as well as gene signatures associated with disorders of pregnancy such as pre-eclampsia. Our findings suggest mechanisms on how optimal immunological interactions between uNK cells and trophoblast enhance reproductive success.
Collapse
Affiliation(s)
- Qian Li
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| | - Andrew Sharkey
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Megan Sheridan
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Elisa Magistrati
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Anna Arutyunyan
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Oisin Huhn
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Carmen Sancho-Serra
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Holly Anderson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Laura Esposito
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ridma Fernando
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Lucy Gardner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Roser Vento-Tormo
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK.
| | | | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
23
|
Rahnavard A, Chatterjee R, Wen H, Gaylord C, Mugusi S, Klatt KC, Smith ER. Molecular epidemiology of pregnancy using omics data: advances, success stories, and challenges. J Transl Med 2024; 22:106. [PMID: 38279125 PMCID: PMC10821542 DOI: 10.1186/s12967-024-04876-7] [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: 02/09/2023] [Accepted: 12/26/2023] [Indexed: 01/28/2024] Open
Abstract
Multi-omics approaches have been successfully applied to investigate pregnancy and health outcomes at a molecular and genetic level in several studies. As omics technologies advance, research areas are open to study further. Here we discuss overall trends and examples of successfully using omics technologies and techniques (e.g., genomics, proteomics, metabolomics, and metagenomics) to investigate the molecular epidemiology of pregnancy. In addition, we outline omics applications and study characteristics of pregnancy for understanding fundamental biology, causal health, and physiological relationships, risk and prediction modeling, diagnostics, and correlations.
Collapse
Affiliation(s)
- Ali Rahnavard
- Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA.
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA.
| | - Ranojoy Chatterjee
- Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
| | - Hui Wen
- Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
| | - Clark Gaylord
- Computational Biology Institute, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA
| | - Sabina Mugusi
- Department of Clinical Pharmacology, Muhimbili University of Health and Allied Sciences, Dar es Salaam, Tanzania
| | - Kevin C Klatt
- Nutritional Sciences & Toxicology, University of California, Berkeley, CA, 94720, USA
| | - Emily R Smith
- Department of Global Health, The Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA.
- Department of Exercise and Nutrition Sciences, The Milken Institute School of Public Health, The George Washington University, Washington, DC, 20052, USA.
| |
Collapse
|
24
|
Garcia-Flores V, Romero R, Tarca AL, Peyvandipour A, Xu Y, Galaz J, Miller D, Chaiworapongsa T, Chaemsaithong P, Berry SM, Awonuga AO, Bryant DR, Pique-Regi R, Gomez-Lopez N. Deciphering maternal-fetal cross-talk in the human placenta during parturition using single-cell RNA sequencing. Sci Transl Med 2024; 16:eadh8335. [PMID: 38198568 PMCID: PMC11238316 DOI: 10.1126/scitranslmed.adh8335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 12/13/2023] [Indexed: 01/12/2024]
Abstract
Labor is a complex physiological process requiring a well-orchestrated dialogue between the mother and fetus. However, the cellular contributions and communications that facilitate maternal-fetal cross-talk in labor have not been fully elucidated. Here, single-cell RNA sequencing (scRNA-seq) was applied to decipher maternal-fetal signaling in the human placenta during term labor. First, a single-cell atlas of the human placenta was established, demonstrating that maternal and fetal cell types underwent changes in transcriptomic activity during labor. Cell types most affected by labor were fetal stromal and maternal decidual cells in the chorioamniotic membranes (CAMs) and maternal and fetal myeloid cells in the placenta. Cell-cell interaction analyses showed that CAM and placental cell types participated in labor-driven maternal and fetal signaling, including the collagen, C-X-C motif ligand (CXCL), tumor necrosis factor (TNF), galectin, and interleukin-6 (IL-6) pathways. Integration of scRNA-seq data with publicly available bulk transcriptomic data showed that placenta-derived scRNA-seq signatures could be monitored in the maternal circulation throughout gestation and in labor. Moreover, comparative analysis revealed that placenta-derived signatures in term labor were mirrored by those in spontaneous preterm labor and birth. Furthermore, we demonstrated that early in gestation, labor-specific, placenta-derived signatures could be detected in the circulation of women destined to undergo spontaneous preterm birth, with either intact or prelabor ruptured membranes. Collectively, our findings provide insight into the maternal-fetal cross-talk of human parturition and suggest that placenta-derived single-cell signatures can aid in the development of noninvasive biomarkers for the prediction of preterm birth.
Collapse
Affiliation(s)
- Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, USA
| | - Adi L Tarca
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, MI 48201, USA
| | - Azam Peyvandipour
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago 8330024, Chile
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Tinnakorn Chaiworapongsa
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Piya Chaemsaithong
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Stanley M Berry
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Awoniyi O Awonuga
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - David R Bryant
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Roger Pique-Regi
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, MD 20892 and Detroit, MI 48201, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
25
|
Saeed H, Wu J, Tesfaye M, Grantz KL, Tekola-Ayele F. Placental accelerated aging in antenatal depression. Am J Obstet Gynecol MFM 2024; 6:101237. [PMID: 38012987 PMCID: PMC10843762 DOI: 10.1016/j.ajogmf.2023.101237] [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: 10/31/2023] [Accepted: 11/22/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Antenatal maternal depression is associated with poor pregnancy outcomes and long-term effects on the offspring. Previous studies have identified links between antenatal depression and placental DNA methylation and between placental epigenetic aging and poor pregnancy outcomes, such as preterm labor and preeclampsia. The relationship between antenatal depression and poor pregnancy outcomes may be partly mediated via placental aging. OBJECTIVE This study aimed to investigate whether antenatal depressive symptoms are associated with placental epigenetic age acceleration, an epigenetic aging clock measure derived from the difference between methylation age and gestational age at delivery. STUDY DESIGN The study included 301 women who provided placenta samples at delivery as part of the Eunice Kennedy Shriver National Institute of Child Health and Human Development Fetal Growth Studies - Singletons that recruited participants from diverse race and ethnic groups at 12 US clinical sites (2009-2013). Women underwent depression screening using the Edinburgh Postnatal Depression Scale up to 6 times across the 3 trimesters of pregnancy. Depressive symptoms status was determined for each pregnancy trimester using an Edinburgh Postnatal Depression Scale score, in which a score of ≥10 was defined as having depressive symptoms and a score of <10 was defined as not having depressive symptoms. Placental DNA methylation was profiled from placenta samples. Placental epigenetic age was estimated using a methylation-based age estimator (placental "epigenetic clock") that has previously been found to have high placental gestational age prediction accuracy for uncomplicated term pregnancies. Placental age acceleration was defined to be the residual upon regressing the estimated epigenetic age on gestational age at delivery. Associations between an Edinburgh Postnatal Depression Scale score of ≥10 and an Edinburgh Postnatal Depression Scale score of <10 in the first, second, and third trimesters of pregnancy (ie, depressive symptoms vs none in each trimester) and placental age acceleration were tested using multivariable linear regression adjusting for maternal age, parity, race and ethnicity, and employment. RESULTS There were 31 (10.3%), 48 (16%), and 49 (16.4%) women with depressive symptoms (ie, Edinburgh Postnatal Depression Scale score of ≥10) in the first, second, and third trimesters of pregnancy, respectively. Of these women, 21 (7.2%) had sustained first- and second-trimester depressive symptoms, 19 (7%) had sustained second- and third-trimester depressive symptoms, and 12 (4.8%) had sustained depressive symptoms throughout pregnancy. Women with depressive symptoms in the second trimester of pregnancy had 0.41 weeks higher placental age acceleration than women without depressive symptoms during the second trimester of pregnancy (β=0.21 weeks [95% confidence interval, -0.17 to 0.58; P=.28] during the first trimester of pregnancy; β=0.41 weeks [95% confidence interval, 0.10-0.71; P=.009] during the second trimester of pregnancy; β=0.17 weeks [95% confidence interval, -0.14 to 0.47; P=.29] during the third trimester of pregnancy). Sustained first- and second-trimester depressive symptoms were associated with 0.72 weeks higher placental age acceleration (95% confidence interval, 0.29-1.15; P=.001) than no depressive symptom in the 2 trimesters. The association between second-trimester depressive symptoms and higher placental epigenetic age acceleration strengthened in the analysis of pregnancies with male fetuses (β=0.53 weeks; 95% confidence interval, 0.06-1.08; P=.03) but was not significant in pregnancies with female fetuses. CONCLUSION Antenatal depressive symptoms during the second trimester of pregnancy were associated with an average of 0.41 weeks of increased placental age acceleration. Accelerated placental aging may play an important role in the underlying mechanism linking antenatal depression to pregnancy complications related to placental dysfunction.
Collapse
Affiliation(s)
- Haleema Saeed
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD (Dr Saeed); Department of Maternal-Fetal Medicine, Medstar Washington Hospital Center, Washington, DC (Drs Saeed)
| | - Jing Wu
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele)
| | - Markos Tesfaye
- Division of Mental Health and Addiction, Norwegian Centre for Mental Disorders Research, University of Oslo, Oslo, Norway (Dr Tesfaye); Department of Psychiatry, St. Paul's Hospital Millennium Medical College, Addis Ababa, Ethiopia (Dr Tesfaye)
| | - Katherine L Grantz
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele)
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD (Drs Wu, Grantz, and Tekola-Ayele).
| |
Collapse
|
26
|
Batorsky R, Ceasrine AM, Shook LL, Kislal S, Bordt EA, Devlin BA, Perlis RH, Slonim DK, Bilbo SD, Edlow AG. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.16.571680. [PMID: 38187648 PMCID: PMC10769274 DOI: 10.1101/2023.12.16.571680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Maternal immune activation is associated with adverse offspring neurodevelopmental outcomes, many mediated by in utero microglial programming. As microglia remain inaccessible throughout development, identification of noninvasive biomarkers reflecting fetal brain microglial programming could permit screening and intervention. We used lineage tracing to demonstrate the shared ontogeny between fetal brain macrophages (microglia) and fetal placental macrophages (Hofbauer cells) in a mouse model of maternal diet-induced obesity, and single-cell RNA-seq to demonstrate shared transcriptional programs. Comparison with human datasets demonstrated conservation of placental resident macrophage signatures between mice and humans. Single-cell RNA-seq identified common alterations in fetal microglial and Hofbauer cell gene expression induced by maternal obesity, as well as sex differences in these alterations. We propose that Hofbauer cells, which are easily accessible at birth, provide novel insights into fetal brain microglial programs, and may facilitate the early identification of offspring vulnerable to neurodevelopmental disorders in the setting of maternal exposures.
Collapse
Affiliation(s)
| | - Alexis M. Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Lydia L. Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evan A. Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin A. Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Roy H. Perlis
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Donna K. Slonim
- Department of Computer Science, Tufts University, Medford, MA
| | - Staci D. Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, MA
| | - Andrea G. Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Owen DM, Kwon M, Huang X, Nagari A, Nandu T, Kraus WL. Genome-wide identification of transcriptional enhancers during human placental development and association with function, differentiation, and disease†. Biol Reprod 2023; 109:965-981. [PMID: 37694817 PMCID: PMC10724456 DOI: 10.1093/biolre/ioad119] [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: 01/28/2023] [Revised: 08/07/2023] [Accepted: 09/09/2023] [Indexed: 09/12/2023] Open
Abstract
The placenta is a dynamic organ that must perform a remarkable variety of functions during its relatively short existence in order to support a developing fetus. These functions include nutrient delivery, gas exchange, waste removal, hormone production, and immune barrier protection. Proper placenta development and function are critical for healthy pregnancy outcomes, but the underlying genomic regulatory events that control this process remain largely unknown. We hypothesized that mapping sites of transcriptional enhancer activity and associated changes in gene expression across gestation in human placenta tissue would identify genomic loci and predicted transcription factor activity related to critical placenta functions. We used a suite of genomic assays [i.e., RNA-sequencing (RNA-seq), Precision run-on-sequencing (PRO-seq), and Chromatin immunoprecipitation-sequencing (ChIP-seq)] and computational pipelines to identify a set of >20 000 enhancers that are active at various time points in gestation. Changes in the activity of these enhancers correlate with changes in gene expression. In addition, some of these enhancers encode risk for adverse pregnancy outcomes. We further show that integrating enhancer activity, transcription factor motif analysis, and transcription factor expression can identify distinct sets of transcription factors predicted to be more active either in early pregnancy or at term. Knockdown of selected identified transcription factors in a trophoblast stem cell culture model altered the expression of key placental marker genes. These observations provide a framework for future mechanistic studies of individual enhancer-transcription factor-target gene interactions and have the potential to inform genetic risk prediction for adverse pregnancy outcomes.
Collapse
Affiliation(s)
- David M Owen
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of General Obstetrics and Gynecology, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Minjung Kwon
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuan Huang
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anusha Nagari
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tulip Nandu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
28
|
Elkin ER, Campbell KA, Lapehn S, Harris SM, Padmanabhan V, Bakulski KM, Paquette AG. Placental single cell transcriptomics: Opportunities for endocrine disrupting chemical toxicology. Mol Cell Endocrinol 2023; 578:112066. [PMID: 37690473 PMCID: PMC10591899 DOI: 10.1016/j.mce.2023.112066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023]
Abstract
The placenta performs essential biologic functions for fetal development throughout pregnancy. Placental dysfunction is at the root of multiple adverse birth outcomes such as intrauterine growth restriction, preeclampsia, and preterm birth. Exposure to endocrine disrupting chemicals during pregnancy can cause placental dysfunction, and many prior human studies have examined molecular changes in bulk placental tissues. Placenta-specific cell types, including cytotrophoblasts, syncytiotrophoblasts, extravillous trophoblasts, and placental resident macrophage Hofbauer cells play unique roles in placental development, structure, and function. Toxicant-induced changes in relative abundance and/or impairment of these cell types likely contribute to placental pathogenesis. Although gene expression insights gained from bulk placental tissue RNA-sequencing data are useful, their interpretation is limited because bulk analysis can mask the effects of a chemical on individual populations of placental cells. Cutting-edge single cell RNA-sequencing technologies are enabling the investigation of placental cell-type specific responses to endocrine disrupting chemicals. Moreover, in situ bioinformatic cell deconvolution enables the estimation of cell type proportions in bulk placental tissue gene expression data. These emerging technologies have tremendous potential to provide novel mechanistic insights in a complex heterogeneous tissue with implications for toxicant contributions to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Elana R Elkin
- School of Public Health, San Diego State University, San Diego, CA, USA.
| | - Kyle A Campbell
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sean M Harris
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, Michigan Medicine, Ann Arbor, MI, USA
| | - Kelly M Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
29
|
Qin L, Wang J, Cheng F, Cheng J, Zhang H, Zheng H, Liu Y, Liang Z, Wang B, Li C, Wang H, Ju Y, Tian H, Meng S. GPC3 and PEG10 peptides associated with placental gp96 elicit specific T cell immunity against hepatocellular carcinoma. Cancer Immunol Immunother 2023; 72:4337-4354. [PMID: 37932427 PMCID: PMC10700408 DOI: 10.1007/s00262-023-03569-2] [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: 06/10/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023]
Abstract
The placenta and tumors can exhibit a shared expression profile of proto-oncogenes. The basis of placenta-derived heat shock protein gp96, which induces prophylactic and therapeutic T cell responses against cancer including hepatocellular carcinoma (HCC), remains unknown. Here, we identified the associated long peptides from human placental gp96 using matrix-assisted laser desorption/ionization-time-of-flight and mass spectrometry and analyzed the achieved proteins through disease enrichment analysis. We found that placental gp96 binds to numerous peptides derived from 73 proteins that could be enriched in multiple cancer types. Epitope-harboring peptides from glypican 3 (GPC3) and paternally expressed gene 10 (PEG10) were the major antigens mediating anti-HCC T cell immunity. Molecular docking analysis showed that the GPC3- and PEG10-derived peptides, mainly obtained from the cytotrophoblast layer of the mature placenta, bind to the lumenal channel and client-bound domain of the gp96 dimer. Immunization with bone marrow-derived dendritic cells pulsed with recombinant gp96-GPC3 or recombinant gp96-PEG10 peptide complex induced specific T cell responses, and T cell transfusion led to pronounced growth inhibition of HCC tumors in nude mice. We demonstrated that the chaperone gp96 can capture antigenic peptides as an efficient approach for defining tumor rejection oncoantigens in the placenta and provide a basis for developing GPC3 and PEG10 peptide-based vaccines against HCC. This study provides insight into the underlying mechanism of the antitumor response mediated by embryonic antigens from fetal tissues, and this will incite more studies to identify potential tumor rejection antigens from placenta.
Collapse
Affiliation(s)
- Lijuan Qin
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiuru Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Fang Cheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiamin Cheng
- Senior Department of Hepatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Han Zhang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Huaguo Zheng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongai Liu
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhentao Liang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Baifeng Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Changfei Li
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Haoyu Wang
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying Ju
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
| | | | - Songdong Meng
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
30
|
Presicce P, Cappelletti M, Morselli M, Ma F, Senthamaraikannan P, Protti G, Nadel BB, Aryan L, Eghbali M, Salwinski L, Pithia N, De Franco E, Miller LA, Pellegrini M, Jobe AH, Chougnet CA, Kallapur SG. Amnion responses to intrauterine inflammation and effects of inhibition of TNF signaling in preterm Rhesus macaque. iScience 2023; 26:108118. [PMID: 37953944 PMCID: PMC10637919 DOI: 10.1016/j.isci.2023.108118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/26/2023] [Accepted: 09/29/2023] [Indexed: 11/14/2023] Open
Abstract
Intrauterine infection/inflammation (IUI) is a frequent complication of pregnancy leading to preterm labor and fetal inflammation. How inflammation is modulated at the maternal-fetal interface is unresolved. We compared transcriptomics of amnion (a fetal tissue in contact with amniotic fluid) in a preterm Rhesus macaque model of IUI induced by lipopolysaccharide with human cohorts of chorioamnionitis. Bulk RNA sequencing (RNA-seq) amnion transcriptomic profiles were remarkably similar in both Rhesus and human subjects and revealed that induction of key labor-mediating genes such as IL1 and IL6 was dependent on nuclear factor κB (NF-κB) signaling and reversed by the anti-tumor necrosis factor (TNF) antibody Adalimumab. Inhibition of collagen biosynthesis by IUI was partially restored by Adalimumab. Interestingly, single-cell transcriptomics, flow cytometry, and immunohistology demonstrated that a subset of amnion mesenchymal cells (AMCs) increase CD14 and other myeloid cell markers during IUI both in the human and Rhesus macaque. Our data suggest that CD14+ AMCs represent activated AMCs at the maternal-fetal interface.
Collapse
Affiliation(s)
- Pietro Presicce
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Monica Cappelletti
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology Medicine at the University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences – Collaboratory at the University of California Los Angeles, Los Angeles, CA, USA
| | - Feiyang Ma
- Department of Molecular, Cell and Developmental Biology Medicine at the University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences – Collaboratory at the University of California Los Angeles, Los Angeles, CA, USA
| | - Paranthaman Senthamaraikannan
- Division of Neonatology/Pulmonary Biology, Cincinnati Children’s Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Giulia Protti
- Institute for Quantitative and Computational Biosciences – Collaboratory at the University of California Los Angeles, Los Angeles, CA, USA
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Brian B. Nadel
- Department of Molecular Cellular and Developmental Biology, and Institute for Genomics and Proteomics, University of California Los Angeles, Los Angeles, CA, USA
- California National Primate Research Center, University of California Davis, Davis, CA, USA
| | - Laila Aryan
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Mansoureh Eghbali
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Lukasz Salwinski
- UCLA-DOE Institute of Genomics and Proteomics, University of California Los Angeles, Los Angeles, CA, USA
| | - Neema Pithia
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Emily De Franco
- Department of Obstetrics/Gynecology, Maternal-Fetal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Lisa A. Miller
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California Davis, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology Medicine at the University of California Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences – Collaboratory at the University of California Los Angeles, Los Angeles, CA, USA
| | - Alan H. Jobe
- Division of Neonatology/Pulmonary Biology, Cincinnati Children’s Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, The University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Claire A. Chougnet
- Division of Immunobiology, Cincinnati Children’s Hospital Research Foundation, and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Suhas G. Kallapur
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
31
|
Xodo S, Londero AP, Orsaria M, Marzinotto S, Colussi G, Cagnacci A, Mariuzzi L, Gri G. Examining the Aryl Hydrocarbon Receptor Network in the Placental Tissues of Pregnancies Complicated by Pre-Eclampsia: An Explorative Case-Control Analysis. Life (Basel) 2023; 13:2122. [PMID: 38004262 PMCID: PMC10672047 DOI: 10.3390/life13112122] [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: 08/14/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Severe maternal and newborn morbidity and mortality associated with pre-eclampsia, which are caused partly by premature delivery, affect a factual proportion of pregnancies. Despite its prevalence, the underlying causes of pre-eclampsia remain elusive, with emerging evidence implicating the aryl hydrocarbon receptor (AhR) in its pathogenesis. This study sought to elucidate the involvement of the AhR and its associated pathway in pre-eclampsia by comparing placental components of the AhR pathway in pregnant individuals with and without pre-eclampsia. This case-control investigation was conducted at the University Hospital of Udine from May 2021 to February 2023. The AhR was assessed using immunohistochemistry and immunofluorescence, and its mRNA was evaluated using a Real-Time Quantitative Reverse Transcription PCR. Levels of mRNA expression were also estimated for other components of the AhR pathway (CYP1B1, IDO1, ARNT, TIPARP, S100A4, and AHRR). Our findings show decreased levels of expression of AhR, IDO1, ARNT, TiPARP, and S100A4 in the placental tissues of individuals with pre-eclampsia compared to controls (p < 0.05). The AhR exhibited a distinct localization within the syncytiotrophoblast (nuclei and cytoplasm) and CD45-positive cells (nuclei and cytoplasm). Furthermore, a significant positive correlation between the AhR and S100A4 (rho = 0.81) was observed in normal placentas, while CYP1B1 displayed a significant negative correlation with the AhR (rho = -0.72), within addition to its negative correlation with TiPARP (rho = -0.83). This study illuminates pre-eclampsia's molecular aberrations, suggesting new diagnostic, therapeutic, and mechanistic approaches. This study emphasizes the need for more research to validate and broaden these findings to improve the management of this complex pregnancy condition.
Collapse
Affiliation(s)
- Serena Xodo
- Clinic of Obstetrics and Gynecology, “Azienda Sanitaria Universitaria Integrata di Udine”, DAME, University of Udine, 33100 Udine, Italy
| | - Ambrogio P. Londero
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Infant Health, University of Genoa, 16132 Genova, Italy;
- Obstetrics and Gynecology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Maria Orsaria
- Institute of Pathology, “Azienda Sanitaria Universitaria Integrata di Udine”, DAME, University of Udine, 33100 Udine, Italy; (M.O.)
| | - Stefania Marzinotto
- Institute of Pathology, “Azienda Sanitaria Universitaria Integrata di Udine”, DAME, University of Udine, 33100 Udine, Italy; (M.O.)
| | - Gianluca Colussi
- Hypertension Unit, Department of Medicine, ASFO “Santa Maria degli Angeli” Hospital of Pordenone, 33170 Pordenone, Italy;
| | - Angelo Cagnacci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Infant Health, University of Genoa, 16132 Genova, Italy;
- Academic Unit of Obstetrics and Gynecology, IRCCS Ospedale San Martino, 16132 Genoa, Italy
| | - Laura Mariuzzi
- Institute of Pathology, “Azienda Sanitaria Universitaria Integrata di Udine”, DAME, University of Udine, 33100 Udine, Italy; (M.O.)
| | - Giorgia Gri
- DIMEC—Department of Medical and Surgical Sciences, University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
32
|
Admati I, Skarbianskis N, Hochgerner H, Ophir O, Weiner Z, Yagel S, Solt I, Zeisel A. Two distinct molecular faces of preeclampsia revealed by single-cell transcriptomics. MED 2023; 4:687-709.e7. [PMID: 37572658 DOI: 10.1016/j.medj.2023.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/04/2023] [Accepted: 07/14/2023] [Indexed: 08/14/2023]
Abstract
INTRODUCTION Preeclampsia is a multisystemic, pregnancy-specific disorder united by new-onset hypertension but with considerable variation in clinical manifestation, onset, and severity. For symptoms to regress, delivery of the placenta is required. For symptoms to regress, delivery of the placenta is required, making the placenta central to preeclampsia pathophysiology. To dissect which placental functions were impacted in two forms of preeclampsia, we studied molecular changes across the cell types of the placenta. METHODS We performed a transcriptomic survey of single-cells and single-nuclei on cases of early- and late-onset preeclampsia with gestation-matched controls. FINDINGS Our data revealed massive dysregulation of gene expression in all cell classes that was almost exclusive to early preeclampsia. For example, an important known receptor/ligand imbalance hallmarking angiogenic disfunction, sFLT1/placental growth factor (PGF), was reflected in striking, cell-autonomous dysregulation of FLT1 and PGF transcription in the syncytium in early preeclampsia only. Stromal cells and vasculature echoed an inflamed, stressed, anti-angiogenic environment. Finally, the placental immune niche set the tone for inflammation in early but not late preeclampsia. Here, fetal-origin Hofbauer and maternal-origin TREM2 macrophages were revealed as surprising main actors, while local cells of the adaptive immune system were largely unaffected. Late preeclampsia showed minimal cellular impact on the placenta. CONCLUSIONS Our survey provides systematic molecular evidence for two distinct diseases. We resolved systematic molecular dysregulation to individual cell types with strong implications for definition, early detection, diagnosis, and treatment. FUNDING Funded by the Preeclampsia Foundation through the Peter Joseph Pappas Research Grant.
Collapse
Affiliation(s)
- Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Niv Skarbianskis
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Hannah Hochgerner
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Osnat Ophir
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Zeev Weiner
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Simcha Yagel
- Division of Obstetrics and Gynecology Hadassah, Hebrew University Medical Centers, Jerusalem, Israel
| | - Ido Solt
- Department of Obstetrics and Gynecology, Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel.
| | - Amit Zeisel
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
33
|
Luo F, Liu F, Guo Y, Xu W, Li Y, Yi J, Fournier T, Degrelle S, Zitouni H, Hernandez I, Liu X, Huang Y, Yue J. Single-cell profiling reveals immune disturbances landscape and HLA-F-mediated immune tolerance at the maternal-fetal interface in preeclampsia. Front Immunol 2023; 14:1234577. [PMID: 37854606 PMCID: PMC10579943 DOI: 10.3389/fimmu.2023.1234577] [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: 06/04/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Background Preeclampsia is a pregnancy-specific disorder that always causes maternal and fetal serious adverse outcome. Disturbances in maternal immune tolerance to embryo at the maternal-fetal interface (MFI) may be associated with preeclampsia onset. Recent studies have revealed the reduced expression pattern of HLA-F at the MFI in preeclampsia, while the mechanism of it mediating maternal fetal immune tolerance has not been revealed. Methods Single-cell RNA sequencing on placental decidua was performed to reveal the immune disturbances landscape at the MFI in preeclampsia. Human Jar cells and NK-92MI cells were employed to study the role of HLA-F in trophoblasts and lymphocyte. Results A total of 101,250 cells were classified into 22 cell clusters. Disease-related IGFBP1+SPP1+ extracellular villus trophoblast (EVT) was identified in the preeclamptic placental decidua, accompanied by newly discovered immune cellular dysfunction such as reduced ribosomal functions of NK populations and abnormal expression of antigen-presenting molecules in most cell clusters. Certain genes that are characteristic of the intermediate stage of myeloid or EVT cell differentiation were found to have unexplored but important functions in the pathogenesis of preeclampsia; specifically, we detected enhanced cell cross-talk between IGFBP1+SPP1+ EVT2 or SPP1+M1 cells and their receptor cell populations at the MFI of PE patients compared to controls. With respect to HLA-F, mIF staining confirmed its reduced expression in PE samples compared to controls. Over-expression of HLA-F in Jar cells promoted cell proliferation, invasion, and migration while under-expression had the opposite effect. In NK-92MI cells, over-expression of HLA-F increased the secretion of immunoregulation cytokines such as CSF1 and CCL22, and promoted adaptive NKG2C+NK cell transformation. Conclusions We revealed the immune disturbance landscape at the MFI in preeclampsia. Our findings regarding cellular heterogeneity and immune cellular dysfunction, as revealed by scRNA-seq, and the function of HLA-F in cells provide new perspectives for further investigation of their roles in the pathogenesis of preeclampsia, and then provide potential new therapeutic target.
Collapse
Affiliation(s)
- Fangyuan Luo
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Fulin Liu
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Yingzhe Guo
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenming Xu
- Department of Obstetrics/Gynecology, Key Laboratory of Obstetric, Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Yilin Li
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
| | - Jun Yi
- Department of Obstetrics and Gynecology Nursing, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
| | - Thierry Fournier
- Pathophysiology & Pharmacotoxicology of the Human Placenta, Pre & Postnatal Microbiota, Université Paris Cité, Paris, France
| | | | - Hedia Zitouni
- Laboratory of Human Genome and Multi-factorial Diseases, Faculty of Pharmacy of Monastir, Monastir, Tunisia
| | - Isabelle Hernandez
- Pathophysiology & Pharmacotoxicology of the Human Placenta, Pre & Postnatal Microbiota, Université Paris Cité, Paris, France
| | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Yu Huang
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
| | - Jun Yue
- Department of Obstetrics and Gynecology, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, University of Electronic Science and Technology, Chengdu, China
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
34
|
Barrozo ER, Seferovic MD, Castro ECC, Major AM, Moorshead DN, Jochum MD, Rojas RF, Shope CD, Aagaard KM. SARS-CoV-2 niches in human placenta revealed by spatial transcriptomics. MED 2023; 4:612-634.e4. [PMID: 37423216 PMCID: PMC10527005 DOI: 10.1016/j.medj.2023.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/21/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Functional placental niches are presumed to spatially separate maternal-fetal antigens and restrict the vertical transmission of pathogens. We hypothesized a high-resolution map of placental transcription could provide direct evidence for niche microenvironments with unique functions and transcription profiles. METHODS We utilized Visium Spatial Transcriptomics paired with H&E staining to generate 17,927 spatial transcriptomes. By integrating these spatial transcriptomes with 273,944 placental single-cell and single-nuclei transcriptomes, we generated an atlas composed of at least 22 subpopulations in the maternal decidua, fetal chorionic villi, and chorioamniotic membranes. FINDINGS Comparisons of placentae from uninfected healthy controls (n = 4) with COVID-19 asymptomatic (n = 4) and symptomatic (n = 5) infected participants demonstrated that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) detection in syncytiotrophoblasts occurred in both the presence and the absence of maternal clinical disease. With spatial transcriptomics, we found that the limit of detection for SARS-CoV-2 was 1/7,000 cells, and placental niches without detectable viral transcripts were unperturbed. In contrast, niches with high SARS-CoV-2 transcript levels were associated with significant upregulation in pro-inflammatory cytokines and interferon-stimulated genes, altered metallopeptidase signaling (TIMP1), with coordinated shifts in macrophage polarization, histiocytic intervillositis, and perivillous fibrin deposition. Fetal sex differences in gene expression responses to SARS-CoV-2 were limited, with confirmed mapping limited to the maternal decidua in males. CONCLUSIONS High-resolution placental transcriptomics with spatial resolution revealed dynamic responses to SARS-CoV-2 in coordinate microenvironments in the absence and presence of clinically evident disease. FUNDING This work was supported by the NIH (R01HD091731 and T32-HD098069), NSF (2208903), the Burroughs Welcome Fund and the March of Dimes Preterm Birth Research Initiatives, and a Career Development Award from the American Society of Gene and Cell Therapy.
Collapse
Affiliation(s)
- Enrico R Barrozo
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Maxim D Seferovic
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Eumenia C C Castro
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Angela M Major
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - David N Moorshead
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, USA; Immunology and Microbiology Graduate Program, Baylor College of Medicine, Houston, TX, USA
| | - Michael D Jochum
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Ricardo Ferral Rojas
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Cynthia D Shope
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
35
|
Huang L, Tu Z, Wei L, Sun W, Wang Y, Bi S, He F, Du L, Chen J, Kzhyshkowska J, Wang H, Chen D, Zhang S. Generating Functional Multicellular Organoids from Human Placenta Villi. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301565. [PMID: 37438660 PMCID: PMC10502861 DOI: 10.1002/advs.202301565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/26/2023] [Indexed: 07/14/2023]
Abstract
The interaction between trophoblasts, stroma cells, and immune cells at the maternal-fetal interface constitutes the functional units of the placenta, which is crucial for successful pregnancy outcomes. However, the investigation of this intricate interplay is restricted due to the absence of efficient experimental models. To address this challenge, a robust, reliable methodology for generating placenta villi organoids (PVOs) from early, late, or diseased pregnancies using air-liquid surface culture is developed. PVOs contain cytotrophoblasts that can self-renew and differentiate directly, along with stromal elements that retain native immune cells. Analysis of scRNA sequencing and WES data reveals that PVOs faithfully recapitulate the cellular components and genetic alterations of the corresponding source tissue. Additionally, PVOs derived from patients with preeclampsia exhibit specific pathological features such as inflammation, antiangiogenic imbalance, and decreased syncytin expression. The PVO-based propagation of primary placenta villi should enable a deeper investigation of placenta development and exploration of the underlying pathogenesis and therapeutics of placenta-originated diseases.
Collapse
Affiliation(s)
- Lijun Huang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Zhaowei Tu
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Liudan Wei
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Wei Sun
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Yifan Wang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Shilei Bi
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Fang He
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Lili Du
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Jingsi Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and ImmunologyMedical Faculty MannheimUniversity of Heidelberg68167MannheimGermany
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health ResearchDepartment of Obstetrics and GynecologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamen361005China
| | - Dunjin Chen
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
- Key Laboratory for Major Obstetric Diseases of Guangdong ProvinceGuangzhou510150China
- Guangdong‐Hong Kong‐Macao Greater Bay Area Higher Education Joint Laboratory of Maternal‐Fetal MedicineGuangzhou510150China
- Guangdong Engineering and Technology Research Center of Maternal‐Fetal MedicineGuangzhou510150China
| | - Shuang Zhang
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhou510150China
| |
Collapse
|
36
|
Moufarrej MN, Quake SR. An inexpensive semi-automated sample processing pipeline for cell-free RNA extraction. Nat Protoc 2023; 18:2772-2793. [PMID: 37567931 DOI: 10.1038/s41596-023-00855-2] [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: 07/06/2022] [Accepted: 05/24/2023] [Indexed: 08/13/2023]
Abstract
Despite advances in automated liquid handling and microfluidics, preparing samples for RNA sequencing at scale generally requires expensive equipment, which is beyond the reach of many academic laboratories. Manual sample preparation remains a slow, expensive and error-prone process. Here, we describe a low-cost, semi-automated pipeline to extract cell-free RNA using one of two commercially available, inexpensive and open-source robotic systems: the Opentrons OT1.0 or OT2.0. Like many RNA isolation protocols, ours can be decomposed into three subparts: RNA extraction, DNA digestion and RNA cleaning and concentration. RT-qPCR data using a synthetic spike-in confirms comparable RNA quality to the gold standard, manual sample processing. The semi-automated pipeline also shows improvement in sample throughput (+12×), time spent (-11×), cost (-3×) and biohazardous waste produced (-4×) compared with its manual counterpart. This protocol enables cell-free RNA extraction from 96 samples simultaneously in 4.5 h; in practice, this dramatically improves the time to results, as we recently demonstrated. Importantly, any laboratory already has most of the parts required (manual pipette and corresponding tips and kits for RNA isolation, cleaning and concentration) to build a semi-automated sample processing pipeline of their own and would only need to purchase or three-dimensionally print a few extra parts (US$5.5 K-12 K in total). This pipeline is also generalizable for many nucleic acid extraction applications, thereby increasing the scale of studies, which can be performed in small research laboratories.
Collapse
Affiliation(s)
- Mira N Moufarrej
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- The Column Group, San Francisco, CA, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Initiative, Redwood City, CA, USA.
| |
Collapse
|
37
|
O'Brien K, Wang Y. The Placenta: A Maternofetal Interface. Annu Rev Nutr 2023; 43:301-325. [PMID: 37603428 DOI: 10.1146/annurev-nutr-061121-085246] [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: 08/23/2023]
Abstract
The placenta is the gatekeeper between the mother and the fetus. Over the first trimester of pregnancy, the fetus is nourished by uterine gland secretions in a process known as histiotrophic nutrition. During the second trimester of pregnancy, placentation has evolved to the point at which nutrients are delivered to the placenta via maternal blood (hemotrophic nutrition). Over gestation, the placenta must adapt to these variable nutrient supplies, to alterations in maternal physiology and blood flow, and to dynamic changes in fetal growth rates. Numerous questions remain about the mechanisms used to transport nutrients to the fetus and the maternal and fetal determinants of this process. Growing data highlight the ability of the placenta to regulate this process. As new technologies and omics approaches are utilized to study this maternofetal interface, greater insight into this unique organ and its impact on fetal development and long-term health has been obtained.
Collapse
Affiliation(s)
- Kimberly O'Brien
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, New York, USA; ,
| | - Yiqin Wang
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, New York, USA; ,
| |
Collapse
|
38
|
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
|
39
|
Zhang B, Xiao W, Qin G, Chen Z, Qiu L, Wang X, Lin Q. Gene loss and co-option of toll-like receptors facilitate paternal immunological adaptation in the brood pouch of pregnant male seahorses. Front Immunol 2023; 14:1224698. [PMID: 37588592 PMCID: PMC10426278 DOI: 10.3389/fimmu.2023.1224698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/04/2023] [Indexed: 08/18/2023] Open
Abstract
Male pregnancy in syngnathids (seahorses, pipefishes, and sea dragons) is an evolutionary innovation in the animal kingdom. Paternal immune resistance to the fetus is a critical challenge, particularly in seahorses with fully enclosed brood pouches and sophisticated placentas. In this study, comparative genomic analysis revealed that all syngnathid species lost three vertebrate-conserved Toll-like receptors (TLR1, TLR2, and TLR9), of which all play essential roles in immune protection and immune tolerance in the uterus and placenta. Quantitative real-time PCR (qRT-PCR) analysis showed that the TLR paralog genes including TLR18, TLR25, and TLR21 were highly expressed in the placenta inside the seahorse brood pouch and changed dynamically during the breeding cycle, suggesting the potentially important role of the TLRs during male pregnancy. Furthermore, the immune challenge test in vitro showed a remarkable expression response from all three TLR genes to specific pathogenic antigens, confirming their immune function in seahorse brood pouches. Notably, the altered antigen recognition spectrum of these genes appeared to functionally compensate in part for the lost TLRs, in contrast to that observed in other species. Therefore, we suggest that gene loss and co-option of TLRs may be a typical evolutionary strategy for facilitating paternal immunological adaptation during male pregnancy.
Collapse
Affiliation(s)
- Bo Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Wanghong Xiao
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
- Sanya Institute of Ocean Eco-Environmental Engineering, Sanya, China
| | - Geng Qin
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
- Sanya Institute of Ocean Eco-Environmental Engineering, Sanya, China
| | - Zelin Chen
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Lihua Qiu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
- Key Laboratory of Aquatic Product Processing, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, China
| | - Xin Wang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| | - Qiang Lin
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
- Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou, China
| |
Collapse
|
40
|
Tabacco S, Ambrosii S, Polsinelli V, Fantasia I, D’Alfonso A, Ludovisi M, Cecconi S, Guido M. Pre-Eclampsia: From Etiology and Molecular Mechanisms to Clinical Tools-A Review of the Literature. Curr Issues Mol Biol 2023; 45:6202-6215. [PMID: 37623210 PMCID: PMC10453909 DOI: 10.3390/cimb45080391] [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: 05/31/2023] [Revised: 07/14/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023] Open
Abstract
Pre-eclampsia is a severe pregnancy-related complication that manifests as a syndrome with multisystem involvement and damage. It has significantly grown in frequency during the past 30 years and could be considered as one of the major causes of maternal and fetal morbidity and mortality. However, the specific etiology and molecular mechanisms of pre-eclampsia are still poorly known and could have a variety of causes, such as altered angiogenesis, inflammations, maternal infections, obesity, metabolic disorders, gestational diabetes, and autoimmune diseases. Perhaps the most promising area under investigation is the imbalance of maternal angiogenic factors and its effects on vascular function, though studies in placental oxidative stress and maternal immune response have demonstrated intriguing findings. However, to determine the relative importance of each cause and the impact of actions aiming to significantly reduce the incidence of this illness, more research is needed. Moreover, it is necessary to better understand the etiologies of each subtype of pre-eclampsia as well as the pathophysiology of other major obstetrical syndromes to identify a clinical tool able to recognize patients at risk of pre-eclampsia early.
Collapse
Affiliation(s)
- Sara Tabacco
- Unit of Obstetrics and Gynecology, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Silvia Ambrosii
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Valentina Polsinelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Ilaria Fantasia
- Unit of Obstetrics and Gynecology, San Salvatore Hospital, 67100 L’Aquila, Italy
| | - Angela D’Alfonso
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Manuela Ludovisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Sandra Cecconi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Maurizio Guido
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
41
|
Sureshchandra S, Doratt BM, True H, Mendoza N, Rincon M, Marshall NE, Messaoudi I. Multimodal profiling of term human decidua demonstrates immune adaptations with pregravid obesity. Cell Rep 2023; 42:112769. [PMID: 37432849 PMCID: PMC10528932 DOI: 10.1016/j.celrep.2023.112769] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/24/2023] [Accepted: 06/23/2023] [Indexed: 07/13/2023] Open
Abstract
Leukocyte diversity of the first-trimester maternal-fetal interface has been extensively described; however, the immunological landscape of the term decidua remains poorly understood. We therefore profiled human leukocytes from term decidua collected via scheduled cesarean delivery. Relative to the first trimester, our analyses show a shift from NK cells and macrophages to T cells and enhanced immune activation. Although circulating and decidual T cells are phenotypically distinct, they demonstrate significant clonotype sharing. We also report significant diversity within decidual macrophages, the frequency of which positively correlates with pregravid maternal body mass index. Interestingly, the ability of decidual macrophages to respond to bacterial ligands is reduced with pregravid obesity, suggestive of skewing toward immunoregulation as a possible mechanism to safeguard the fetus against excessive maternal inflammation. These findings are a resource for future studies investigating pathological conditions that compromise fetal health and reproductive success.
Collapse
Affiliation(s)
- Suhas Sureshchandra
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| | - Brianna M Doratt
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA
| | - Heather True
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA; Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Norma Mendoza
- Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA
| | - Monica Rincon
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Nicole E Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Ilhem Messaoudi
- Institute for Immunology, University of California, Irvine, Irvine, CA 92697, USA; Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
42
|
Ricci CA, Reid DM, Sun J, Santillan DA, Santillan MK, Phillips NR, Goulopoulou S. Maternal and fetal mitochondrial gene dysregulation in hypertensive disorders of pregnancy. Physiol Genomics 2023; 55:275-285. [PMID: 37184228 PMCID: PMC10292966 DOI: 10.1152/physiolgenomics.00005.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/16/2023] Open
Abstract
Mitochondrial dysfunction has been implicated in pregnancy-induced hypertension (PIH). The role of mitochondrial gene dysregulation in PIH, and consequences for maternal-fetal interactions, remain elusive. Here, we investigated mitochondrial gene expression and dysregulation in maternal and placental tissues from pregnancies with and without PIH; further, we measured circulating mitochondrial DNA (mtDNA) mutational load, an index of mtDNA integrity. Differential gene expression analysis followed by Time Course Gene Set Analysis (TcGSA) was conducted on publicly available high throughput sequencing transcriptomic data sets. Mutational load analysis was carried out on peripheral mononuclear blood cells from healthy pregnant individuals and individuals with preeclampsia. Thirty mitochondrial differentially expressed genes (mtDEGs) were detected in the maternal cell-free circulating transcriptome, whereas nine were detected in placental transcriptome from pregnancies with PIH. In PIH pregnancies, maternal mitochondrial dysregulation was associated with pathways involved in inflammation, cell death/survival, and placental development, whereas fetal mitochondrial dysregulation was associated with increased production of extracellular vesicles (EVs) at term. Mothers with preeclampsia did not exhibit a significantly different degree of mtDNA mutational load. Our findings support the involvement of maternal mitochondrial dysregulation in the pathophysiology of PIH and suggest that mitochondria may mediate maternal-fetal interactions during healthy pregnancy.NEW & NOTEWORTHY This study identifies aberrant maternal and fetal expression of mitochondrial genes in pregnancies with gestational hypertension and preeclampsia. Mitochondrial gene dysregulation may be a common etiological factor contributing to the development of de novo hypertension in pregnancy-associated hypertensive disorders.
Collapse
Affiliation(s)
- Contessa A Ricci
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Danielle M Reid
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Jie Sun
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Donna A Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Mark K Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States
| | - Nicole R Phillips
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, Texas, United States
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas, United States
- Department of Gynecology and Obstetrics, Lawrence D. Longo MD Center for Perinatal Biology, Loma Linda University School of Medicine, Loma Linda, California, United States
| |
Collapse
|
43
|
Hui L, Ellis K, Mayen D, Pertile MD, Reimers R, Sun L, Vermeesch J, Vora NL, Chitty LS. Position statement from the International Society for Prenatal Diagnosis on the use of non-invasive prenatal testing for the detection of fetal chromosomal conditions in singleton pregnancies. Prenat Diagn 2023; 43:814-828. [PMID: 37076973 DOI: 10.1002/pd.6357] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/15/2023] [Indexed: 04/21/2023]
Abstract
Key points
What is already known about this topic?
In 2015, the International Society for Prenatal Diagnosis (ISPD) published its first position statement on the use of non‐invasive prenatal testing (NIPT) to screen for aneuploidy. Widespread uptake across the globe and subsequent published research has shed new light on test performance and implementation issues.
What does this study add?
This new position statement replaces the 2015 statement with updated information on the current technologies, clinical experience, and implementation practices.
As an international organization, ISPD recognizes that there are important population‐specific considerations in the organization of prenatal screening and diagnosis. These opinions are designed to apply to high income settings where prenatal screening for aneuploidy is an established part of antenatal care.
This position statement is not a clinical practice guideline but represents the consensus opinion of the current ISPD Board based on the current state of knowledge and clinical practice.
Collapse
Affiliation(s)
- Lisa Hui
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Reproductive Epidemiology Group, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Perinatal Medicine, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Department of Obstetrics and Gynaecology, Northern Health, Epping, Victoria, Australia
| | - Katie Ellis
- Illumina ANZ, Sydney, New South Wales, Australia
| | - Dora Mayen
- Genetics Clinic, Hospital Angeles Lomas, Estado de Mexico, Mexico
| | - Mark D Pertile
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Rebecca Reimers
- San Diego Perinatal Center, Rady Children's Hospital, San Diego, California, USA
- Scripps Research Institute, La Jolla, California, USA
| | - Luming Sun
- Department of Fetal Medicine & Prenatal Diagnosis Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | | | - Neeta L Vora
- Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Division of Maternal Fetal Medicine, Chapel Hill, North Carolina, USA
| | - Lyn S Chitty
- Great Ormond Street NHS Foundation Trust, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
44
|
Yang H, Chelariu-Raicu A, Makri D, Ori C, Ribeiro PCP, Kolben T, Kuhn C, Kessler M, Schulz C, Mahner S, Jeschke U, von Schönfeldt V. Updates of placental macrophages: Origins, molecular markers, functions, and related diseases. J Reprod Immunol 2023; 157:103942. [PMID: 36989681 DOI: 10.1016/j.jri.2023.103942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
Placental macrophages are highly heterogeneous cells with differential phenotypes and functions defined by differential origins and modulated by the changing placental environment. During pregnancy, placental macrophages play a critical role in embryo implantation, placenta formation and homeostasis, fetal development and parturition. This review summarizes recent findings on the cellular origin of placental macrophages, and provide a comprehensive description of their phenotypes, corresponding molecular markers and functions in human placenta. Finally, alterations of placental macrophages in pregnancy-related diseases are discussed.
Collapse
|
45
|
Yang J, Gong L, Liu Q, Zhao H, Wang Z, Li X, Tian W, Zhou Q. Single-cell RNA-seq reveals developmental deficiencies in both the placentation and the decidualization in women with late-onset preeclampsia. Front Immunol 2023; 14:1142273. [PMID: 37283740 PMCID: PMC10239844 DOI: 10.3389/fimmu.2023.1142273] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/21/2023] [Indexed: 06/08/2023] Open
Abstract
Preeclampsia (PE) is a leading cause of maternal and fetal morbidity and mortality. Although increasing lines of evidence suggest that both the placenta and the decidua likely play roles in the pathogenesis of PE, the molecular mechanism of PE remains elusive partly because of the heterogeneity nature of the maternal-fetal interface. In this study, we perform single-cell RNA-seq on the placenta and the decidual from patients with late-onset PE (LOPE) and women in normal pregnancy. Analyses of single-cell transcriptomes reveal that in LOPE, there are likely a global development deficiency of trophoblasts with impaired invasion of extravillous trophoblasts (EVT) and increased maternal immune rejection and inflammation in the placenta, while there are likely insufficient decidualization of decidual stromal cells (DSC), increased inflammation, and suppressed regulatory functions of decidual immune cells. These findings improve our understanding of the molecular mechanisms of PE.
Collapse
Affiliation(s)
- Jing Yang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, Department of Computational Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Lili Gong
- Obstetrics and Gynaecology Hospital, Fudan University, Shanghai, China
| | - Qiming Liu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, Department of Computational Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Huanqiang Zhao
- Obstetrics and Gynaecology Hospital, Fudan University, Shanghai, China
| | - Zekun Wang
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, Department of Computational Biology, School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaotian Li
- Obstetrics and Gynaecology Hospital, Fudan University, Shanghai, China
- Obstetrics and Gynecology Hospital, Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| | - Weidong Tian
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, Department of Computational Biology, School of Life Sciences, Fudan University, Shanghai, China
- Children’s Hospital of Fudan University, Shanghai, China
- Children’s Hospital of Shandong University, Jinan, Shandong, China
| | - Qiongjie Zhou
- Obstetrics and Gynaecology Hospital, Fudan University, Shanghai, China
- Obstetrics and Gynecology Hospital, Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
46
|
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
|
47
|
Liu X, Aneas I, Sakabe N, Anderson RL, Billstrand C, Paz C, Kaur H, Furner B, Choi S, Prichina AY, Enninga EAL, Dong H, Murtha A, Crawford GE, Kessler JA, Grobman W, Nobrega MA, Rana S, Ober C. Single cell profiling at the maternal-fetal interface reveals a deficiency of PD-L1 + non-immune cells in human spontaneous preterm labor. Sci Rep 2023; 13:7903. [PMID: 37193763 DOI: 10.1038/s41598-023-35051-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 05/11/2023] [Indexed: 05/18/2023] Open
Abstract
The mechanisms that underlie the timing of labor in humans are largely unknown. In most pregnancies, labor is initiated at term (≥ 37 weeks gestation), but in a signifiicant number of women spontaneous labor occurs preterm and is associated with increased perinatal mortality and morbidity. The objective of this study was to characterize the cells at the maternal-fetal interface (MFI) in term and preterm pregnancies in both the laboring and non-laboring state in Black women, who have among the highest preterm birth rates in the U.S. Using mass cytometry to obtain high-dimensional single-cell resolution, we identified 31 cell populations at the MFI, including 25 immune cell types and six non-immune cell types. Among the immune cells, maternal PD1+ CD8 T cell subsets were less abundant in term laboring compared to term non-laboring women. Among the non-immune cells, PD-L1+ maternal (stromal) and fetal (extravillous trophoblast) cells were less abundant in preterm laboring compared to term laboring women. Consistent with these observations, the expression of CD274, the gene encoding PD-L1, was significantly depressed and less responsive to fetal signaling molecules in cultured mesenchymal stromal cells from the decidua of preterm compared to term women. Overall, these results suggest that the PD1/PD-L1 pathway at the MFI may perturb the delicate balance between immune tolerance and rejection and contribute to the onset of spontaneous preterm labor.
Collapse
Affiliation(s)
- Xiao Liu
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Ivy Aneas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Noboru Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | | | | | - Cristina Paz
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Harjot Kaur
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Brian Furner
- Center for Research Informatics, University of Chicago, Chicago, IL, USA
| | - Seong Choi
- Center for Research Informatics, University of Chicago, Chicago, IL, USA
| | | | | | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Amy Murtha
- Department of Obstetrics and Gynecology, Duke University Health Systems, Durham, NC, USA
- Rutgers RWJ Medical School, New Brunswick, NJ, USA
| | - Gregory E Crawford
- Department of Pediatrics and Center for Genomics and Computational Biology, Duke University, Durham, NC, USA
| | - John A Kessler
- Department of Neurology and Institute for Stem Cell Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - William Grobman
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Marcelo A Nobrega
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Sarosh Rana
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
48
|
Liu Z, Yu Y, Zhang X, Wang C, Pei J, Gu W. Transcriptomic profiling in hypoxia-induced trophoblast cells for preeclampsia. Placenta 2023; 136:8-17. [PMID: 37001424 DOI: 10.1016/j.placenta.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/24/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023]
Abstract
This study aimed to identify the expression profile of mRNAs and analyze the associated pathways in hypoxia-induced trophoblast cells to understand the effect of hypoxia on the pathophysiology of preeclampsia (PE). We downloaded two gene expression datasets (GSE47187 and GSE60432) from the Gene Expression Omnibus (GEO) datasets to identify altered transcriptomes. GEO2R, gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and protein-protein interaction (PPI) networks were used to reveal the functional roles and regulatory networks of the differentially expressed genes (DEGs). In total, 224 DEGs (91 upregulated and 133 downregulated) were identified, and the "HIF-1 signaling pathway" was activated in placentas from patients with PE. We validated the expression levels of five proteins in the plasma of NP and PE patients during early or late pregnancy using western blotting. In primary trophoblast cells cultured under hypoxic conditions, 754 DEGs were identified, including 362 upregulated and 392 downregulated genes. These DEGs were associated with the "HIF-1signaling pathway," "response to hypoxia," and several glucose metabolism pathways. In addition, a PPI network was constructed, and an important module, including 18 hub genes, was identified. Finally, we validated 18 hub genes using qRT-PCR. Furthermore, we performed microarray profiling of hypoxia-treated HTR8/SVneo cells (immortalized human first-trimester extravillous trophoblast cells) to validate the DEGs and pathways identified in hypoxia-induced primary trophoblast cells. Our results stress the differential expression profiles of mRNAs in hypoxia-induced trophoblast cells, which provide potential pathophysiological mechanisms for preeclampsia.
Collapse
|
49
|
Jin H, Zhang Y, Fan Z, Wang X, Rui C, Xing S, Dong H, Wang Q, Tao F, Zhu Y. Identification of novel cell-free RNAs in maternal plasma as preterm biomarkers in combination with placental RNA profiles. J Transl Med 2023; 21:256. [PMID: 37046301 PMCID: PMC10100253 DOI: 10.1186/s12967-023-04083-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/25/2023] [Indexed: 04/14/2023] Open
Abstract
BACKGROUND Preterm birth (PTB) is the main driver of newborn deaths. The identification of pregnancies at risk of PTB remains challenging, as the incomplete understanding of molecular mechanisms associated with PTB. Although several transcriptome studies have been done on the placenta and plasma from PTB women, a comprehensive description of the RNA profiles from plasma and placenta associated with PTB remains lacking. METHODS Candidate markers with consistent trends in the placenta and plasma were identified by implementing differential expression analysis using placental tissue and maternal plasma RNA-seq datasets, and then validated by RT-qPCR in an independent cohort. In combination with bioinformatics analysis tools, we set up two protein-protein interaction networks of the significant PTB-related modules. The support vector machine (SVM) model was used to verify the prediction potential of cell free RNAs (cfRNAs) in plasma for PTB and late PTB. RESULTS We identified 15 genes with consistent regulatory trends in placenta and plasma of PTB while the full term birth (FTB) acts as a control. Subsequently, we verified seven cfRNAs in an independent cohort by RT-qPCR in maternal plasma. The cfRNA ARHGEF28 showed consistence in the experimental validation and performed excellently in prediction of PTB in the model. The AUC achieved 0.990 for whole PTB and 0.986 for late PTB. CONCLUSIONS In a comparison of PTB versus FTB, the combined investigation of placental and plasma RNA profiles has shown a further understanding of the mechanism of PTB. Then, the cfRNA identified has the capacity of predicting whole PTB and late PTB.
Collapse
Affiliation(s)
- Heyue Jin
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
| | - Yimin Zhang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China
| | - Zhigang Fan
- Department of Neonatology, Ma'anshan Maternal and Child Health Hospital, Ma'anshan, Anhui, China
| | - Xianyan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, Anhui, China
| | - Chen Rui
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, Anhui, China
| | - Shaozhen Xing
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Hongmei Dong
- Department of Obstetrics, Ma'anshan Maternal and Child Health Hospital, Ma'anshan, Anhui, China
| | - Qunan Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
- Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, Anhui, China.
| | - Fangbiao Tao
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.
| | - Yumin Zhu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China.
- MOE Key Laboratory of Population Health Across Life Cycle, No 81 Meishan Road, Hefei, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
50
|
Jiang X, Zhai J, Xiao Z, Wu X, Zhang D, Wan H, Xu Y, Qi L, Wang M, Yu D, Liu Y, Wu H, Sun R, Xia S, Yu K, Guo J, Wang H. Identifying a dynamic transcriptomic landscape of the cynomolgus macaque placenta during pregnancy at single-cell resolution. Dev Cell 2023; 58:806-821.e7. [PMID: 37054708 DOI: 10.1016/j.devcel.2023.03.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 01/10/2023] [Accepted: 03/16/2023] [Indexed: 04/15/2023]
Abstract
Supporting healthy pregnancy outcomes requires a comprehensive understanding of the cellular hierarchy and underlying molecular mechanisms in the primate placenta during gestation. Here, we present a single-cell transcriptome-wide view of the cynomolgus macaque placenta throughout gestation. Bioinformatics analyses and multiple validation experiments suggested that placental trophoblast cells exhibited stage-specific differences across gestation. Interactions between trophoblast cells and decidual cells also showed gestational stage-dependent differences. The trajectories of the villous core cells indicated that placental mesenchymal cells were derived from extraembryonic mesoderm (ExE.Meso) 1, whereas placental Hofbauer cells, erythrocytes, and endothelial cells were derived from ExE.Meso2. Comparative analyses of human and macaque placentas uncovered conserved features of placentation across species, and the discrepancies of extravillous trophoblast cells (EVTs) between human and macaque correlated to their differences in invasion patterns and maternal-fetal interactions. Our study provides a groundwork for elucidating the cellular basis of primate placentation.
Collapse
Affiliation(s)
- Xiangxiang Jiang
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, China
| | - Jinglei Zhai
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zhenyu Xiao
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; School of Life Science, Beijing Institute of Technology, Beijing 100081, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Xulun Wu
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Dan Zhang
- Key Laboratory of Reproductive Genetics (Ministry of Education), Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Haifeng Wan
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanhong Xu
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Luqing Qi
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Meijiao Wang
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Dainan Yu
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yawei Liu
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Hao Wu
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Run Sun
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Shuwei Xia
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Kunyuan Yu
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Jingtao Guo
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Hongmei Wang
- The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China.
| |
Collapse
|