1
|
Wong FC, Kim CE, Garcia-Alonso L, Vento-Tormo R. The human endometrium: atlases, models, and prospects. Curr Opin Genet Dev 2025; 92:102341. [PMID: 40154154 DOI: 10.1016/j.gde.2025.102341] [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: 01/31/2025] [Accepted: 02/26/2025] [Indexed: 04/01/2025]
Abstract
Approximately every month, the human endometrium undergoes a cycle of proliferation, differentiation, and, in the absence of pregnancy, shedding and repair. Each cycle relies on intricate interorgan coordination of hormonal secretions. Endometrial dysfunction causes significant health complications, including abnormal menstrual bleeding and endometriosis. However, effective diagnosis and treatments are hampered by understudied aetiology. Recent single-cell profiling has disentangled the diverse and dynamic nature of the endometrium, revealing regulatory roles of WNT, NOTCH, and TGFβ signalling. These insights have informed mechanistic studies enabled by advanced in vitro models that capture endometrial cellular heterogeneity and structure. In this review, we outline key single-cell transcriptomics atlases and models that provided new avenues for studying endometrial biology, discuss their limitations, and propose future directions.
Collapse
Affiliation(s)
- Frederick Ck Wong
- Wellcome Sanger Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | | | - Luz Garcia-Alonso
- Wellcome Sanger Institute, Cambridge, UK. https://twitter.com/@LuzGarAl
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Cambridge, UK; Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
2
|
Keenen MM, Yang L, Liang H, Farmer VJ, Worota RE, Singh R, Gladfelter AS, Coyne CB. Comparative analysis of the syncytiotrophoblast in placenta tissue and trophoblast organoids using snRNA sequencing. eLife 2025; 13:RP101170. [PMID: 40424181 DOI: 10.7554/elife.101170] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025] Open
Abstract
The syncytiotrophoblast (STB) is a multinucleated cell layer that forms the outer surface of human chorionic villi. Its unusual structure, with billions of nuclei in a single cell, makes it difficult to resolve using conventional single-cell methods. To better understand STB differentiation, we performed single-nucleus and single-cell RNA sequencing on placental tissue and trophoblast organoids (TOs). Single-nucleus RNA-seq was essential for capturing STB populations, revealing three nuclear subtypes: a juvenile subtype co-expressing CTB and STB markers, one enriched in oxygen sensing genes, and another in transport and GTPase signaling. Organoids grown in suspension culture (STBout) showed higher expression of STB markers, hormones, and a greater proportion of the transport-associated nuclear subtype while TOs grown with an inverted polarity (STBin) exhibited a higher proportion of the oxygen sensing nuclear subtype. Gene regulatory analysis identified conserved STB markers, including the chromatin remodeler RYBP. Although RYBP knockout did not impair fusion, it downregulated CSH1 and upregulated oxygen-sensing genes. Comparing STB expression in first trimester, term, and TOs revealed shared features but context-dependent variability. These findings establish TOs as a robust platform to model STB differentiation and nuclear heterogeneity, providing insight into the regulatory networks that shape placental development and function.
Collapse
Affiliation(s)
- Madeline M Keenen
- Duke University School of Medicine, Department of Cell Biology, Durham, United States
| | - Liheng Yang
- Duke University School of Medicine, Department of Integrative Immunobiology, Durham, United States
| | - Huan Liang
- Duke University School of Medicine, Department of Cell Biology, Durham, United States
- Duke University School of Medicine, Department of Biostatistics and Bioinformatics, Durham, United States
| | - Veronica J Farmer
- Duke University School of Medicine, Department of Cell Biology, Durham, United States
| | - Rizban E Worota
- Duke University School of Medicine, Department of Integrative Immunobiology, Durham, United States
| | - Rohit Singh
- Duke University School of Medicine, Department of Cell Biology, Durham, United States
- Duke University School of Medicine, Department of Biostatistics and Bioinformatics, Durham, United States
| | - Amy S Gladfelter
- Duke University School of Medicine, Department of Cell Biology, Durham, United States
| | - Carolyn B Coyne
- Duke University School of Medicine, Department of Integrative Immunobiology, Durham, United States
- Duke Human Vaccine Institute, Durham, United States
| |
Collapse
|
3
|
Presicce P, Morselli M, Jeong A, Altendahl M, Martinez G, Del Vecchio G, Devaskar SU, Pellegrini M, Afshar Y, Kallapur SG. Placental Privilege: Evidence of organ resilience in severe COVID-19 in pregnancy. Placenta 2025:S0143-4004(25)00156-0. [PMID: 40383676 DOI: 10.1016/j.placenta.2025.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/27/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND COVID-19 infection in pregnancy is associated with preterm birth and an increased risk of severe disease, needing intensive care admission for management of maternal multi-organ failure. The placenta, a fetal organ, functions as a barrier at the maternal interface and expresses the SARS-CoV-2 viral receptors. However, placental infection and transplacental transfer of virus are rare, suggesting placental resistance to viral infection. Here, we seek to determine the impact of severe COVID-19 infection on maternal, newborn, and placental outcomes. METHODS A prospectively recruited cohort of pregnant COVID-19 patients (n = 204) at a quaternary perinatal academic center were retrospectively analyzed. During pregnancy umbilical artery (UA) Doppler assessment was performed to assess placental function. At delivery, maternal and fetal outcomes were assessed, with paired maternal peripheral blood and placenta samples collected (n = 26) for bulk RNA sequencing (RNA-seq). Post-sequencing analysis with single cell deconvolution and pathway analysis was performed. RESULTS Maternally-indicated preterm births were more frequent in severe, but not asymptomatic or mild/moderate COVID-19 infection. In severe COVID-19 infection, UA Doppler assessment was normal. Rates of fetal growth restriction and placenta:birth weight ratios were similar between groups. RNA-seq showed a distinct adaptive immune activation signature in peripheral blood while placental transcripts showed no significant changes in immune cell types. CONCLUSION Despite multi-organ failure, severe COVID-19 did not significantly impact placental function and transcriptomics with iatrogenic preterm birth indicated for maternal-indications.
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
| | - Marco Morselli
- Department of Chemistry, Life Sciences and Environmental Sustainability (S.C.V.S.A.), University of Parma, Parma, Italy
| | - Anhyo Jeong
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Marie Altendahl
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Guadalupe Martinez
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Giorgia Del Vecchio
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA
| | - Sherin U Devaskar
- Divisions of Neonatology and Developmental Biology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, 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; Molecular Biology Institute, University of California, Los Angeles, USA
| | - Yalda Afshar
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles, 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
|
4
|
Duan H, Deng W, Kzhyshkowska J, Chen D, Zhang S. Macrophage at maternal-fetal Interface: Perspective on pregnancy and related disorders. Placenta 2025:S0143-4004(25)00158-4. [PMID: 40399151 DOI: 10.1016/j.placenta.2025.05.005] [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: 01/20/2025] [Revised: 04/13/2025] [Accepted: 05/08/2025] [Indexed: 05/23/2025]
Abstract
Immune cells at the maternal-fetal interface (MFI) undergo dynamic changes to facilitate fetal growth and development during pregnancy. In contrast to the adaptive immune system, where effector T cells, Tregs, and suppressor T cells play key roles in maintaining immune tolerance toward the semi-allogeneic fetus, the innate immune system-comprising decidual nature killer (dNK) cells, macrophages, and dendritic cells (DCs)-makes up a significant portion of the decidual leukocyte population. These innate immune cells are crucial in modulating trophoblast invasion, spiral artery remodeling, and apoptotic cell phagocytosis. Dysregulation of the innate immune system has been linked to impaired uterine vessel remodeling and defective trophoblast invasion, which can lead to complications such as spontaneous abortion, preeclampsia (PE), and preterm. This review focuses on recent advancements in understanding the innate immune defenses at the maternal-fetal interface and their connections to pregnancy-related diseases, with particular emphasis on the role of macrophages.
Collapse
Affiliation(s)
- Haoran Duan
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Weinan Deng
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany; German Red Cross Blood Service Baden- Württemberg-Hessen, 68167, Mannheim, Germany; Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
| | - Dunjin Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| | - Shuang Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China; Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China.
| |
Collapse
|
5
|
Nonn O, Debnath O, Valdes DS, Sallinger K, Secener AK, Fischer C, Tiesmeyer S, Nimo J, Kuenzer T, Ulrich J, Maxian T, Knöfler M, Karau P, Bartolomaeus H, Kroneis T, Frolova A, Neuper L, Haase N, Malt A, Müller-Bötticher N, Kräker K, Kedziora S, Forstner D, Eils R, Schmidt-Ullrich R, Haider S, Verlohren S, Stern C, Sugulle M, Jones S, Thilaganathan B, Kaitu’u-Lino TJ, Tong S, Huppertz B, El-Heliebi A, Staff AC, Coscia F, Müller DN, Dechend R, Gauster M, Ishaque N, Herse F. Senescent Syncytiotrophoblast Secretion During Early Onset Preeclampsia. Hypertension 2025; 82:787-799. [PMID: 39440423 PMCID: PMC12002046 DOI: 10.1161/hypertensionaha.124.23362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Preeclampsia is a severe hypertensive disorder in pregnancy that causes preterm delivery, maternal and fetal morbidity, mortality, and life-long sequelae. Understanding the pathogenesis of preeclampsia is a critical first step toward protecting mother and child from this syndrome and increased risk of cardiovascular disease later in life. However, effective early predictive tests and therapies for preeclampsia are scarce. METHODS To identify novel markers and signaling pathways for early onset preeclampsia, we profiled human maternal-fetal interface units (fetal villi and maternal decidua) from early onset preeclampsia and healthy controls using single-nucleus RNA sequencing combined with spatial transcriptomics. The placental syncytiotrophoblast is in direct contact with maternal blood and forms the barrier between fetal and maternal circulation. RESULTS We identified different transcriptomic states of the endocrine syncytiotrophoblast nuclei with patterns of dysregulation associated with a senescence-associated secretory phenotype and a spatial dysregulation of senescence in the placental trophoblast layer. Elevated senescence markers were validated in placental tissues of clinical multicenter cohorts. Importantly, several secreted senescence-associated secretory phenotype factors were elevated in maternal blood already in the first trimester. We verified the secreted senescence markers, PAI-1 (plasminogen activator inhibitor 1) and activin A, as identified in our single-nucleus RNA sequencing model as predictive markers before clinical preeclampsia diagnosis. CONCLUSIONS This indicates that increased syncytiotrophoblast senescence appears weeks before clinical manifestation of early onset preeclampsia, suggesting that the dysregulated preeclamptic placenta starts with higher cell maturation resulting in premature and increased senescence-associated secretory phenotype release. These senescence-associated secretory phenotype markers may serve as an additional early diagnostic tool for this syndrome.
Collapse
Affiliation(s)
- Olivia Nonn
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Olivia Debnath
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Daniela S. Valdes
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Katja Sallinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
- Centre for Biomarker Research in Medicine, Graz, Austria (K.S., T. Kroneis, A.E.-H.)
| | - Ali Kerim Secener
- Max Delbrück Center - Berlin Institute for Medical Systems Biology (MDC-BIMSB), Berlin, Germany (A.K.S., C.F.)
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (A.K.S.)
| | - Cornelius Fischer
- Max Delbrück Center - Berlin Institute for Medical Systems Biology (MDC-BIMSB), Berlin, Germany (A.K.S., C.F.)
| | - Sebastian Tiesmeyer
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Jose Nimo
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| | - Thomas Kuenzer
- Institute for Medical Informatics, Statistics and Documentation (T. Kuenzer), Medical University of Graz, Austria
| | - Juliane Ulrich
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| | - Theresa Maxian
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Austria (T.M., M.K., S.H.)
| | - Martin Knöfler
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Austria (T.M., M.K., S.H.)
| | - Philipp Karau
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Hendrik Bartolomaeus
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Thomas Kroneis
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
- Centre for Biomarker Research in Medicine, Graz, Austria (K.S., T. Kroneis, A.E.-H.)
| | - Alina Frolova
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- Institute of Molecular Biology and Genetic of the National Academy of Sciences of Ukraine (NASU), Kyiv, Ukraine (A.F.)
| | - Lena Neuper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Nadine Haase
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Alexander Malt
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Niklas Müller-Bötticher
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Kristin Kräker
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Sarah Kedziora
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Roland Eils
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Ruth Schmidt-Ullrich
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| | - Sandra Haider
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Austria (T.M., M.K., S.H.)
| | - Stefan Verlohren
- Department of Obstetrics and Gynaecology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (S.V.)
| | - Christina Stern
- Department of Obstetrics and Gynaecology, University Hospital Graz, Medical University Graz, Austria (C.S.)
| | - Meryam Sugulle
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway (M.S., A.C.S.)
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (M.S., A.C.S.)
| | - Stuart Jones
- Clinical Biochemistry, King George’s Hospital, London, United Kingdom (S.J.)
| | - Basky Thilaganathan
- Fetal Medicine Unit, St George’s University Hospitals NHS Foundation Trust, London, United Kingdom (B.T.)
| | - Tu’uhevaha J. Kaitu’u-Lino
- Department of Obstetrics and Gynaecology (Mercy Hospital for Women), The University of Melbourne, VIC, Australia (T.J.K.-L., S. Tong)
| | - Stephen Tong
- Department of Obstetrics and Gynaecology (Mercy Hospital for Women), The University of Melbourne, VIC, Australia (T.J.K.-L., S. Tong)
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Amin El-Heliebi
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
- Centre for Biomarker Research in Medicine, Graz, Austria (K.S., T. Kroneis, A.E.-H.)
| | - Anne Cathrine Staff
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway (M.S., A.C.S.)
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (M.S., A.C.S.)
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| | - Dominik N. Müller
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Ralf Dechend
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
- Department of Cardiology and Nephrology, HELIOS Clinic, Berlin, Germany (R.D.)
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Naveed Ishaque
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Florian Herse
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| |
Collapse
|
6
|
Vaughan OR, Maksym K, Hillman S, Spencer RN, Hristova M, David AL, Lange S. Placental Protein Citrullination Signatures Are Modified in Early- and Late-Onset Fetal Growth Restriction. Int J Mol Sci 2025; 26:4247. [PMID: 40362485 PMCID: PMC12071715 DOI: 10.3390/ijms26094247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Fetal growth restriction (FGR) is an obstetric condition most frequently caused by placental dysfunction. It is a major cause of perinatal morbidity with limited treatment options, so identifying the underpinning mechanisms is important. Peptidylarginine deiminases (PADs) are calcium-activated enzymes that mediate post-translational citrullination (deimination) of proteins, through conversion of arginine to citrulline. Protein citrullination leads to irreversible changes in protein structure and function and is implicated in many pathobiological processes. Whether placental protein citrullination occurs in FGR is poorly understood. We assessed protein citrullination and PAD isozyme abundance (PAD1, 2, 3, 4 and 6) in human placental samples from pregnancies complicated by early- and late-onset FGR, compared to appropriate-for-gestational-age (AGA) controls. Proteomic mass spectrometry demonstrated that the placental citrullinome profile changed in both early- and late-onset FGR, with 112 and 345 uniquely citrullinated proteins identified in early- and late-onset samples, respectively. Forty-four proteins were citrullinated only in control AGA placentas. The proteins that were uniquely citrullinated in FGR placentas were enriched for gene ontology (GO) terms related to neurological, developmental, immune and metabolic pathways. A greater number of GO and human phenotype pathways were functionally enriched for citrullinated proteins in late- compared with early-onset FGR. Correspondingly, late-onset but not early-onset FGR was associated with significantly increased placental abundance of PAD2 and citrullinated histone H3, determined by Western blotting. PAD3 was downregulated in early-onset FGR while abundance of PAD 1, 4 and 6 was less altered in FGR. Our findings show that placental protein citrullination is altered in FGR placentas, potentially contributing to the pathobiology of placental dysfunction.
Collapse
Affiliation(s)
- Owen R. Vaughan
- Department of Maternal and Fetal Medicine, EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (O.R.V.); (S.H.); (R.N.S.); (A.L.D.)
| | - Kasia Maksym
- Women’s Health Division, University College London Hospitals NHS Foundation Trust, London NW1 2PG, UK;
| | - Sara Hillman
- Department of Maternal and Fetal Medicine, EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (O.R.V.); (S.H.); (R.N.S.); (A.L.D.)
| | - Rebecca N. Spencer
- Department of Maternal and Fetal Medicine, EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (O.R.V.); (S.H.); (R.N.S.); (A.L.D.)
- Department of Obstetrics and Gynaecology, University of Leeds, Leeds LS2 9JT, UK
| | - Mariya Hristova
- Department of Neonatology, EGA Institute for Women’s Health, University College London, London WC1E 6BT, UK;
| | - Anna L. David
- Department of Maternal and Fetal Medicine, EGA Institute for Women’s Health, University College London, London WC1E 6HX, UK; (O.R.V.); (S.H.); (R.N.S.); (A.L.D.)
| | - Sigrun Lange
- Department of Neonatology, EGA Institute for Women’s Health, University College London, London WC1E 6BT, UK;
- Pathobiology and Extracellular Vesicles Research Group, School of Life Sciences, University of Westminster, London W1W 6UW, UK
| |
Collapse
|
7
|
Velagala S, Phan L, Eke C, Fernandes A, Rice TA, Olaloye O, Konnikova L. Spatial single-cell analysis identifies placental villi structuraland immune remodeling across gestation. Mucosal Immunol 2025:S1933-0219(25)00043-1. [PMID: 40288579 DOI: 10.1016/j.mucimm.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 02/04/2025] [Accepted: 04/13/2025] [Indexed: 04/29/2025]
Abstract
Pregnancies rely upon the balance between fetal and maternal immune systems. Employing imaging mass cytometry, this study creates a spatial map landscape to unravel the cellular dynamics within the placental villi (PV). Consistent with previous data we report structural remodeling in PV, highlighted by increased syncytial trophoblasts, vascular smooth muscle cells, and endothelial cells in term PV. Additionally, we identified that rare immune populations including dendritic cells, innate lymphoid cells, and myeloid-derived suppressor cells in mid-gestation were more abundant and activated than in term placentas, suggesting a potentially tolerogenic state. Conversely, various macrophage subtypes were increased and in combination with rare T-cells exhibited heightened activation markers, possibly indicating increased signaling in preparation for parturition. In mid-gestation, fibroblasts had increased interactions with trophoblasts, while term samples exhibited close adjacencies between trophoblasts, vascular smooth muscle cells, and macrophages. This study offers insights into the PV cellular composition changes between mid and full-term samples, providing a foundation for future studies to understand the mechanisms of preterm birth and other pregnancy complications.
Collapse
Affiliation(s)
| | - Long Phan
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Chino Eke
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Analie Fernandes
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Tyler A Rice
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | | | - Liza Konnikova
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA; Program in Human and Translational Immunology, Yale School of Medicine, New Haven, CT, USA; Program in Translational Biomedicine, Yale School of Medicine, New Haven, CT, USA; Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
8
|
Wu G, Liang Y, Xi Q, Zuo Y. New Insights and Implications of Cell-Cell Interactions in Developmental Biology. Int J Mol Sci 2025; 26:3997. [PMID: 40362237 PMCID: PMC12072105 DOI: 10.3390/ijms26093997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
The dynamic and meticulously regulated networks established the foundation for embryonic development, where the intercellular interactions and signal transduction assumed a pivotal role. In recent years, high-throughput technologies such as single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) have advanced dramatically, empowering the systematic dissection of cell-to-cell regulatory networks. The emergence of comprehensive databases and analytical frameworks has further provided unprecedented insights into embryonic development and cell-cell interactions (CCIs). This paper reviewed the exponential increased CCIs works related to developmental biology from 2008 to 2023, comprehensively collected and categorized 93 analytical tools and 39 databases, and demonstrated its practical utility through illustrative case studies. In parallel, the article critically scrutinized the persistent challenges within this field, such as the intricacies of spatial localization and transmembrane state validation at single-cell resolution, and underscored the interpretative limitations inherent in current analytical frameworks. The development of CCIs' analysis tools with harmonizing multi-omics data and the construction of cross-species dynamically updated CCIs databases will be the main direction of future research. Future investigations into CCIs are poised to expeditiously drive the application and clinical translation within developmental biology, unlocking novel dimensions for exploration and progress.
Collapse
Affiliation(s)
| | | | | | - Yongchun Zuo
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010021, China; (G.W.); (Y.L.); (Q.X.)
| |
Collapse
|
9
|
de Alwis N, Beard S, Baird L, Binder NK, Pritchard N, Tong S, Kaitu'u-Lino TJ, Hui L, Hannan NJ. Phosphoglucomutase 5 gene transcripts are expressed by the human placenta and differentially regulated in placental dysfunction. Sci Rep 2025; 15:11381. [PMID: 40180976 PMCID: PMC11968825 DOI: 10.1038/s41598-025-94498-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
The placenta plays an essential role facilitating nutrient, gas and waste exchange between the maternal and fetal systems for optimal fetal growth. When placental development is impaired and the placenta dysfunctional, serious pregnancy complications such as fetal growth restriction and preeclampsia may arise. Previously, phosphoglucomutase-5 (PGM5) transcripts were found to be highly elevated in the blood of patients whose pregnancies were complicated by fetal growth restriction and preeclampsia. As both conditions feature placental insufficiency, here we aimed to characterise PGM5 levels in the healthy and dysfunctional placenta. PGM5 expression was detectable in all placental samples across gestation, in cases of preterm preeclampsia, fetal growth restriction and controls. PGM5 mRNA expression was significantly downregulated in the pathological placentas compared to controls, but PGM5 protein production was not dysregulated. Isolated cytotrophoblast and placental explant tissue exposed to hypoxia (modelling placental dysfunction) demonstrated significantly increased PGM5 expression, but again did not change protein levels. Silencing PGM5 expression under hypoxic conditions in primary cytotrophoblast did not alter anti-angiogenic sFLT-1 secretion but increased expression of multiple genes associated with cell growth, apoptosis and oxidative stress, whilst also increasing cell viability. Expression of PGM5 in all placental samples assessed suggests that PGM5 has functions in the placenta. However, further investigation could be performed to explore the discrepancies in protein and mRNA expression, as well as the precise function of PGM5 in the placenta, and whether altered PGM5 levels may be important for placental development.
Collapse
Affiliation(s)
- Natasha de Alwis
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Sally Beard
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Lydia Baird
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Natalie K Binder
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Natasha Pritchard
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Stephen Tong
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
| | - Lisa Hui
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia
- Northern Health, Epping, Melbourne, VIC, 3076, Australia
| | - Natalie J Hannan
- Department of Obstetrics, Gynaecology and Newborn Health, The University of Melbourne and Mercy Hospital for Women, 163 Studley Rd, Heidelberg, Melbourne, VIC, 3084, Australia.
- Northern Health, Epping, Melbourne, VIC, 3076, Australia.
| |
Collapse
|
10
|
Solt I, Cohen SM, Admati I, Beharier O, Dominsky O, Yagel S. Placenta at single-cell resolution in early and late preeclampsia: insights and clinical implications. Am J Obstet Gynecol 2025; 232:S176-S189. [PMID: 40253080 DOI: 10.1016/j.ajog.2025.01.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 04/21/2025]
Abstract
Preeclampsia, one of the great obstetrical syndromes, manifests through diverse maternal and fetal complications and remains a leading contributor to adverse perinatal outcomes. In this review, we describe our work on single-cell and single-nuclei RNA sequencing to elucidate the molecular mechanisms that underlie early- and late-onset preeclampsia. Analysis of 46 cell types, encompassing approximately 90,000 cells from placental tissues collected after delivery, demonstrated cellular dysregulation in early-onset preeclampsia, whereas late-onset preeclampsia showed comparatively subtle changes. These findings were observed in all cell lines, including all types of trophoblast, lymphoid, myeloid, stromal, and endothelial cells. Key findings in early-onset preeclampsia included disrupted syncytiotrophoblast and extravillous trophoblast angiogenic signaling, characterized by an up-regulation of FLT1 and down-regulation of PGF, consistent with an angiogenic imbalance. The stromal and vascular compartments exhibited stress-induced transcriptomic shifts. Both endothelial cells and pericytes showed evidence of stress, including up-regulation of heat shock proteins and markers of apoptosis. In addition, the inflammation- and stress-responsive states were more abundant in early-onset preeclampsia than in matched controls. Inflammatory pathways were markedly up-regulated in both the maternal and fetal immune cells; for example, we observed a marked increase in pro-inflammatory cytokines, including secreted phosphoprotein 1 and C-X-C motif chemokine ligand 2 and 3. Conversely, late-onset preeclampsia retained adaptive placental features with localized dysregulation of extracellular matrix remodeling and angiogenic markers, underscoring its possible maternal cardiovascular etiology. Single-cell and single-nuclei RNA sequencing investigations of placental tissues support the proposed classification of preeclampsia into a placental dysfunction type, primarily presenting early in pregnancy, and a maternal cardiovascular maladaptation type, primarily presenting later in pregnancy, each with distinct biomarkers, risk factors, and therapeutic targets. The early-onset preeclampsia findings advocate for interventions that target angiogenic pathways, such as RNA-based therapies that target specific cells of the placenta, to modulate soluble fms-like tyrosine kinase-1 levels. In contrast, late-onset preeclampsia management may benefit from maternal cardiovascular optimization, including individualized antihypertensive and metabolic treatments. These results underscore the heterogeneity of preeclampsia, emphasizing the need for individualized diagnostic and therapeutic strategies. This molecular atlas of preeclampsia advances our understanding of the complex interplay among elements of the maternal-placental-fetal array, thereby bridging clinical phenotypes and cellular mechanisms. Future research should focus on integrating these insights into longitudinal studies to develop precision medicine approaches for preeclampsia to enhance outcomes for mothers and neonates.
Collapse
Affiliation(s)
- Ido Solt
- Department of Obstetrics and Gynecology, Rambam Health Care Campus & Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Sarah M Cohen
- Division of Obstetrics and Gynecology, Hadassah Hebrew University Medical Centers, Jerusalem, Israel
| | - Inbal Admati
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology Haifa, Israel
| | - Ofer Beharier
- Division of Obstetrics and Gynecology, Hadassah Hebrew University Medical Centers, Jerusalem, Israel
| | - Omri Dominsky
- Department of Obstetrics and Gynecology, Lis Hospital for Women's Health Sourasky Medical Center, affiliated with the Faculty of Medicine at Tel Aviv University, Tel Aviv, Israel
| | - Simcha Yagel
- Division of Obstetrics and Gynecology, Hadassah Hebrew University Medical Centers, Jerusalem, Israel.
| |
Collapse
|
11
|
Levenson D, Romero R, Miller D, Galaz J, Garcia-Flores V, Neshek B, Pique-Regi R, Gomez-Lopez N. The maternal-fetal interface at single-cell resolution: uncovering the cellular anatomy of the placenta and decidua. Am J Obstet Gynecol 2025; 232:S55-S79. [PMID: 40253083 DOI: 10.1016/j.ajog.2024.12.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 12/12/2024] [Accepted: 12/27/2024] [Indexed: 04/21/2025]
Abstract
The maternal-fetal interface represents a critical site of immunological interactions that can greatly influence pregnancy outcomes. The unique cellular composition and cell-cell interactions taking place within these tissues has spurred substantial research efforts focused on the maternal-fetal interface. With the recent advent of single-cell technologies, multiple investigators have applied such methods to gain an unprecedented level of insight into maternal-fetal communication. Here, we provide an overview of the dynamic cellular composition and cell-cell communications at the maternal-fetal interface as reported by single-cell investigations. By primarily focusing on data from pregnancies in the second and third trimesters, we aim to showcase how single-cell technologies have bolstered the foundational understanding of each cell's contribution to physiologic gestation. Indeed, single-cell technologies have enabled the examination of classical placental cells, such as the trophoblast, as well as uncovered new roles for structural cells now recognized as active participants in pregnancy and parturition, such as decidual and fetal stromal cells, which are reviewed herein. Furthermore, single-cell data investigating the ontogeny, function, differentiation, and interactions among immune cells present at the maternal-fetal interface, namely macrophages, T cells, dendritic cells, neutrophils, mast cells, innate lymphoid cells, natural killer cells, and B cells are discussed in this review. Moreover, a key output of single-cell investigations is the inference of cell-cell interactions, which has been leveraged to not only dissect the intercellular communications within specific tissues but also between compartments such as the decidua basalis and placental villi. Collectively, this review emphasizes the ways by which single-cell technologies have expanded the understanding of cell composition and cellular processes underlying pregnancy in mid-to-late gestation at the maternal-fetal interface, which can prompt their continued application to reveal new pathways and targets for the treatment of obstetrical disease.
Collapse
Affiliation(s)
- Dustyn Levenson
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO; Department of Physiology, 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 (NICHD/NIH/DHHS), Bethesda, MD; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| | - Derek Miller
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Jose Galaz
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Valeria Garcia-Flores
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Barbara Neshek
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI
| | - Nardhy Gomez-Lopez
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO.
| |
Collapse
|
12
|
Xu J, He X, Zhang S, Li L, Li P. Expression of co-signaling molecules TIM-3/Galectin-9 at the maternal-fetal interface. Placenta 2025; 163:43-50. [PMID: 40068377 DOI: 10.1016/j.placenta.2025.03.002] [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: 11/26/2024] [Revised: 01/31/2025] [Accepted: 03/02/2025] [Indexed: 04/01/2025]
Abstract
INTRODUCTION During early pregnancy, fetal placental tissue implants into maternal decidual tissue, forming a unique interface where maternal immune cells do not reject the invading fetal cells. Given the roles of Galectin-9 and Tim-3 in tumor immune regulation, studying their distribution and function at this interface may provide insights into recurrent pregnancy loss. METHODS This study uses single-cell transcriptomics, spatial transcriptomics, and multiplex immunohistochemistry to examine the expression and localization of Galectin-9 and TIM-3. Hormone-induced decidualization of immortalized human endometrial stromal cells was conducted to investigate Galectin-9 expression. RESULTS The major immune cells in the maternal decidua, such as T cells, NK cells, and macrophages, co-express Galectin-9 and TIM-3. Unlike TIM-3, Galectin-9 is also highly expressed in endothelial cells and decidualized stromal cells. Among placenta-derived cells, Hofbauer cells (HBs) and Placenta-associated maternal monocytes/macrophages (PAMMs) exhibit high expression of both Galectin-9 and TIM-3, while trophoblast cells show relatively low levels of expression. Additionally, hormone-induced decidualization significantly upregulates Galectin-9 expression in endometrial stromal cells. DISCUSSION The research results suggest that Galectin-9 and TIM-3, as important immune co-signaling molecules, may play a crucial role in maintaining the immune-tolerant microenvironment at the maternal-fetal interface. Additionally, the association between decidualization and Galectin-9 expression reveals its potential role in pregnancy maintenance, providing new insights for the study of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Jingliang Xu
- Sichuan Jinxin Xinan Women's and Children's Hospital, Chengdu, China
| | - Xuqing He
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Sujuan Zhang
- Sichuan Jinxin Xinan Women's and Children's Hospital, Chengdu, China
| | - Li Li
- Sichuan Jinxin Xinan Women's and Children's Hospital, Chengdu, China.
| | - Penghao Li
- Sichuan Jinxin Xinan Women's and Children's Hospital, Chengdu, China; Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China; Yunnan Jinxin Jiuzhou Hospital, Yunnan, China.
| |
Collapse
|
13
|
Sousa A, Besong OTO, Wachman EM, Taglauer ES, Beane JE, Kefella Y, Koo JS, Saia K, Jones HE, Zhang H. Placental transcriptome analysis in opioid-exposed versus non-opioid exposed pregnancies. Placenta 2025; 162:27-34. [PMID: 39983471 PMCID: PMC11908891 DOI: 10.1016/j.placenta.2025.02.012] [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: 12/01/2024] [Revised: 02/05/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
INTRODUCTION Opioid exposure during pregnancy may significantly alter gene expression in the placenta, potentially disrupting its function and influencing fetal brain development. These alterations may contribute to adverse outcomes such as neonatal opioid withdrawal syndrome (NOWS). In this study, we aim to systematically investigate the changes in placental gene expression associated with maternal opioid exposure to better understand the underlying molecular mechanisms and their implications for fetal health. METHODS Fresh placental tissue samples were collected from 18 opioid-exposed pregnancies and 26 non-opioid-exposed control pregnancies. Transcriptomic changes related to opioid exposure were assessed using RNA sequencing (RNA-seq). RESULTS Among the 16,172 genes detected, 55 showed differential expression (Padjusted < 0.25 or Punadjusted < 0.001) in opioid-exposed placentas. Gene Set Enrichment Analysis (GSEA) revealed that the differentially expressed genes were primarily associated with immune responses, neuronal development and function, as well as cell replication and division. Computational deconvolution using the PlacentaCellEnrich program identified significant enrichment of upregulated genes in decidual NK cells. Furthermore, integrative analysis of DNA methylation and gene expression showed an enrichment of differentially methylated genes among downregulated genes in opioid-exposed placentas. DISCUSSION Our findings suggest that opioid exposure during pregnancy may disrupt critical placental pathways, particularly those involved in immune responses. Future studies focusing on transcriptomic changes in specific placental cell types will be essential for fully understanding the structural and functional alterations in the placenta due to opioid exposure during pregnancy.
Collapse
Affiliation(s)
- Aneya Sousa
- Department of Pediatrics, Boston Medical Center, Boston, MA, USA
| | - Ojong Tabi Ojong Besong
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Elisha M Wachman
- Department of Pediatrics, Boston Medical Center, Boston, MA, USA.
| | | | - Jennifer E Beane
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Yohana Kefella
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ji Sun Koo
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kelley Saia
- Department of Obstetrics and Gynecology, Boston Medical Center, Boston, MA, USA
| | - Hendree E Jones
- Department of Obstetrics & Gynecology, University of North Carolina, Chapel Hill, NC, USA
| | - Huiping Zhang
- Department of Psychiatry, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Section of Biomedical Genetics, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| |
Collapse
|
14
|
Unjitwattana T, Huang Q, Yang Y, Tao L, Yang Y, Zhou M, Du Y, Garmire LX. Single-cell RNA-seq data have prevalent blood contamination but can be rescued by Originator, a computational tool separating single-cell RNA-seq by genetic and contextual information. Genome Biol 2025; 26:52. [PMID: 40069819 PMCID: PMC11895284 DOI: 10.1186/s13059-025-03495-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 02/05/2025] [Indexed: 03/15/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) data from complex human tissues have prevalent blood cell contamination during the sample preparation process. They may also comprise cells of different genetic makeups. We propose a new computational framework, Originator, which deciphers single cells by genetic origin and separates immune cells of blood contamination from those of expected tissue-resident cells. We demonstrate the accuracy of Originator at separating immune cells from the blood and tissue as well as cells of different genetic origins, using a variety of artificially mixed and real datasets, including pancreatic cancer and placentas as examples.
Collapse
Affiliation(s)
- Thatchayut Unjitwattana
- Department of Biomedical Engineering, University of Michigan, 2200 , Bonisteel, Ann Arbor, MI, 48109, USA
| | - Qianhui Huang
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave, Ann Arbor, MI, 48109, USA
| | - Yiwen Yang
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave, Ann Arbor, MI, 48109, USA
| | - Leyang Tao
- Department of Biomedical Engineering, University of Michigan, 2200 , Bonisteel, Ann Arbor, MI, 48109, USA
| | - Youqi Yang
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Mengtian Zhou
- Department of Statistics, University of Michigan, 1085 S University Ave, Ann Arbor, MI, 48109, USA
| | - Yuheng Du
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave, Ann Arbor, MI, 48109, USA
| | - Lana X Garmire
- Department of Biomedical Engineering, University of Michigan, 2200 , Bonisteel, Ann Arbor, MI, 48109, USA.
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave, Ann Arbor, MI, 48109, USA.
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
15
|
Shen Z, Zhao Y, Xu X, Yang H, He S, Ma J, Zhang S, Hou P, Sui F. Single-cell RNA sequencing integrated with bulk RNA sequencing analysis of clock circadian regulator with prognostic and immune microenvironment in thyroid cancer. Transl Oncol 2025; 53:102299. [PMID: 39892222 PMCID: PMC11833347 DOI: 10.1016/j.tranon.2025.102299] [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: 06/26/2024] [Revised: 01/05/2025] [Accepted: 01/28/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND Disruption of circadian rhythm was found to be associated with immune infiltration and thyroid cancer. However, the role of clock circadian regulator (CLOCK) in the progression of thyroid cancer and its immune microenvironment remains largely unexplored. Therefore, our aim was to explore the role and potential mechanism of CLOCK in thyroid cancer. METHODS Single cell sequencing analysis and bulk RNA sequencing analysis was used for LASSO regression and Kaplan-Meier survival estimates. Potential mechanism analysis were gained through KEGG/GO analysis, GSEA analysis and PPI network. In vivo and in vitro experiment was used for further validation. RESULTS The result showed CLOCK protein was overexpressed in thyroid cancer compared with normal tissue in both thyroid specific mouse model and human sample. A prognostic model incorporating CLOCK and other related genes (FAT4, OR6K2, STK40, TMEM63A, HRCT1, SUPT5H, and OR2C3) was developed using LASSO regression. Functional assay and bioinformatics analysis indicated that CLOCK knockdown hindered tumor growth and the activity of MAPK signaling. Besides, analyses of gene enrichment, signaling pathways, and immune checkpoints suggested that CLOCK might inhibit immune infiltration within the tumor microenvironment. Confirmatory in vitro experiments and immunohistochemical assays in human samples further linked high CLOCK expression to reduced T cell cytotoxicity and infiltration. CONCLUSION These findings underscore the pivotal role of CLOCK in thyroid cancer prognosis and immune suppression, highlighting its potential as a target for therapeutic intervention and prognostic assessment in thyroid cancer management.
Collapse
Affiliation(s)
- Zhen Shen
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Yuelei Zhao
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Xinxin Xu
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Huini Yang
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Shuting He
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Junchi Ma
- School of Information Engineering, Chang'an University, Shaanxi Province, PR China
| | - Shaoqiang Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China; International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, Xi'an 710061, Shaanxi Province, PR China
| | - Fang Sui
- Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, PR China.
| |
Collapse
|
16
|
Veličković M, Kadam L, Kim J, Zemaitis KJ, Veličković D, Gao Y, Wu R, Fillmore TL, Orton D, Williams SM, Monroe ME, Moore RJ, Piehowski PD, Bramer LM, Myatt L, Burnum-Johnson KE. Advanced multi-modal mass spectrometry imaging reveals functional differences of placental villous compartments at microscale resolution. Nat Commun 2025; 16:2061. [PMID: 40021619 PMCID: PMC11871073 DOI: 10.1038/s41467-025-57107-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/12/2025] [Indexed: 03/03/2025] Open
Abstract
The placenta is a complex and heterogeneous organ that links the mother and fetus, playing a crucial role in nourishing and protecting the fetus throughout pregnancy. Integrative spatial multi-omics approaches can provide a systems-level understanding of molecular changes underlying the mechanisms leading to the histological variations of the placenta during healthy pregnancy and pregnancy complications. Herein, we advance our metabolome-informed proteome imaging (MIPI) workflow to include lipidomic imaging, while also expanding the molecular coverage of metabolomic imaging by incorporating on-tissue chemical derivatization (OTCD). The improved MIPI workflow advances biomedical investigations by leveraging state-of-the-art molecular imaging technologies. Lipidome imaging identifies molecular differences between two morphologically distinct compartments of a placental villous functional unit, syncytiotrophoblast (STB) and villous core. Next, our advanced metabolome imaging maps villous functional units with enriched metabolomic activities related to steroid and lipid metabolism, outlining distinct molecular distributions across morphologically different villous compartments. Complementary proteome imaging on these villous functional units reveals a plethora of fatty acid- and steroid-related enzymes uniquely distributed in STB and villous core compartments. Integration across our advanced MIPI imaging modalities enables the reconstruction of active biological pathways of molecular synthesis and maternal-fetal signaling across morphologically distinct placental villous compartments with micrometer-scale resolution.
Collapse
Affiliation(s)
- Marija Veličković
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Leena Kadam
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA
| | - Joonhoon Kim
- Energy and Environment Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Kevin J Zemaitis
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Dušan Veličković
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Yuqian Gao
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ruonan Wu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Thomas L Fillmore
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Daniel Orton
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Sarah M Williams
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Matthew E Monroe
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Paul D Piehowski
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lisa M Bramer
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| | - Leslie Myatt
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, OR, USA.
| | - Kristin E Burnum-Johnson
- The Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
17
|
Keenen MM, Yang L, Liang H, Farmer VJ, Worota RE, Singh R, Gladfelter AS, Coyne CB. Comparative analysis of the syncytiotrophoblast in placenta tissue and trophoblast organoids using snRNA sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.01.601571. [PMID: 39005304 PMCID: PMC11244908 DOI: 10.1101/2024.07.01.601571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The outer surface of chorionic villi in the human placenta consists of a single multinucleated cell called the syncytiotrophoblast (STB). The unique cellular ultrastructure of the STB presents challenges in deciphering its gene expression signature at the single-cell level, as the STB contains billions of nuclei in a single cell. There are many gaps in understanding the molecular mechanisms and developmental trajectories involved in STB formation and differentiation. To identify the underlying control of the STB, we performed comparative single nucleus (SN) and single cell (SC) RNA sequencing on placental tissue and tissue-derived trophoblast organoids (TOs). We found that SN RNA sequencing was essential to capture the STB population from both tissue and TOs. Differential gene expression and pseudotime analysis of TO-derived STB identified three distinct nuclear subtypes reminiscent of those recently identified in vivo . These included a juvenile nuclear population that exhibited both CTB and STB marker expression, a population enriched in genes involved in oxygen sensing, and finally a subtype enriched in transport and GTPase signaling molecules. Notably, suspension culture conditions of TOs that restore the native orientation of the STB (STB out ) showed elevated expression of canonical STB markers and pregnancy hormones, along with a greater proportion of the STB nucleus subtype specializing in transport and GTPase signaling, compared to those cultivated with an inverted STB polarity (STB in ). Gene regulatory analysis identified novel markers of STB differentiation conserved in tissue and TOs, including the chromatin remodeler RYBP, that exhibited STB-specific RNA and protein expression. CRISPR/Cas9 knockout of RYBP in STB in TOs did not impact cell-cell fusion; however, bulk RNA sequencing revealed downregulation of the pregnancy hormone CSH1 and upregulation of multiple genes associated with the oxygen-sensing STB nuclear subtype. Finally, we compared STB gene expression signatures amongst first trimester tissue, full-term tissue, and TOs, identifying many commonalities but also notable variability across each sample type. This indicates that STB gene expression is responsive to its environmental context. Our findings emphasize the utility of TOs to accurately model STB differentiation and the distinct nuclear subtypes observed in vivo , offering a versatile platform for unraveling the molecular mechanisms governing STB functions in placental biology and disease.
Collapse
|
18
|
Lee J, Lee NK, Moon JH. Gestational Diabetes Mellitus: Mechanisms Underlying Maternal and Fetal Complications. Endocrinol Metab (Seoul) 2025; 40:10-25. [PMID: 39844628 PMCID: PMC11898322 DOI: 10.3803/enm.2024.2264] [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: 12/03/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Gestational diabetes mellitus (GDM) affects over 10% of all pregnancies, both in Korea and worldwide. GDM not only increases the risk of adverse pregnancy outcomes such as preeclampsia, preterm birth, macrosomia, neonatal hypoglycemia, and shoulder dystocia, but it also significantly increases the risk of developing postpartum type 2 diabetes mellitus and cardiovascular disease in the mother. Additionally, GDM is linked to a higher risk of childhood obesity and diabetes in offspring, as well as neurodevelopmental disorders, including autistic spectrum disorder. This review offers a comprehensive summary of clinical epidemiological studies concerning maternal and fetal complications and explores mechanistic investigations that reveal the underlying pathophysiology.
Collapse
Affiliation(s)
- Jooyeop Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Armed Forces Yangju Hospital, Yangju, Korea
| | - Na Keum Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Joon Ho Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
19
|
Llorca T, Ruiz-Magaña MJ, Abadía AC, Ruiz-Ruiz C, Olivares EG. Decidual stromal cells: fibroblasts specialized in immunoregulation during pregnancy. Trends Immunol 2025; 46:138-152. [PMID: 39947975 DOI: 10.1016/j.it.2024.12.007] [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: 07/18/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025]
Abstract
Decidual stromal cells (DSCs) are involved in immunoregulatory mechanisms that prevent fetal rejection by the mammalian maternal immune system. Recent studies using single-cell RNA sequencing demonstrated the existence of different types of human and mouse DSCs, highlighting corresponding differentiation (decidualization) pathways, and suggesting their involvement in the immune response during normal and pathological pregnancy. DSCs may be considered tissue-specialized fibroblasts because both DSCs and fibroblasts share phenotypic and functional similarities in immunologically challenged tissues, especially in terms of their immune functions. Indeed, fibroblasts can setup, support, and suppress immune responses and these functions are also performed by DSCs. Moreover, fibroblasts and DSCs can induce ectopic foci as tertiary lymphoid structures (TLSs), and endometriosis, respectively. Thus, understanding DSC immunoregulatory functions is of timely relevance.
Collapse
Affiliation(s)
- Tatiana Llorca
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain
| | - María José Ruiz-Magaña
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Biología Celular, Universidad de Granada, Granada, Spain.
| | - Ana C Abadía
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Carmen Ruiz-Ruiz
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain
| | - Enrique G Olivares
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, Armilla, Granada, Spain; Departamento de Bioquímica y Biología Molecular III e Inmunología, Universidad de Granada, Granada, Spain.
| |
Collapse
|
20
|
Li P, Zeng L, Yan X, Zhu Z, Gu Q, He X, Zhang S, Mao R, Xu J, Xie F, Wang H, Li Z, Shu J, Zhang W, Sha Y, Huang J, Su M, Zheng Q, Ma J, Zhou X, Li M, Pan H, Li Y, Yan M, Chen X, Li M, Long K, Kong F, Tang C, Huang J, Su C, Li J, Fang Z, Chen M, Tian E, Zhong Y, Gu J. Molecular and cellular morphology of placenta unveils new mechanisms of reproductive immunology. J Adv Res 2025:S2090-1232(25)00044-X. [PMID: 39842636 DOI: 10.1016/j.jare.2025.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
INTRODUCTION Despite of numerous studies of the placenta, some molecular and cellular characteristics, particularly the relationship among different cell types, have not been well understood. We aim to investigate the basic and intricate details of cellular and molecular elements in early and late phase placentas to gain better understanding of the immune regulation of human reproductive process. METHODS A novel combination of techniques of spatial transcriptomics(ST), multiple immunohistochemistry, and a dual labeling combining immunohistochemistry and (fluorescence in situ hybridization) FISH on normal and ectopic pregnancy and animal models was employed to investigate the placenta at tissue, cell, protein and molecular levels and to trace the fetal and maternal origin of every cell in early and late placentas. RESULTS Original discoveries include early expression of immune checkpoint proteins in embryo trophoblasts even before implantation. The detailed distributional relationships among different cell types of fetal and maternal origins in placenta and decidua indicate an immune rejection of the mother towards the fetus and this was counterbalanced by immune inhibitory proteins and blocking antibody Immunoglobulin G4 (IgG4) at the junction between the fetus and the mother. In contrary to common believe, we found that vascular endothelial and glandular epithelial cells in the decidua remain maternal in origin and were not replaced by fetal cells. At term placenta, fetal immune cells infiltrated into the maternal side of the decidus and vice versa indicating a possible immune reaction between fetal and maternal immune systems and suggesting a possible immune mechanism for trigger of parturition. The ability of trophoblasts to create an immune suppressed environment was also supported by findings in ectopic pregnancy and the animal models. CONCLUSION The findings indicate a fetus-driven mechanism of immune balance involving both cellular and humoral immunity in human reproduction.
Collapse
Affiliation(s)
- Penghao Li
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China; Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Liting Zeng
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China; Shenshan Medical Center, Memorial Hospital of Sun Yat-sen University, Shanwei 516600, China
| | - Xiaomiao Yan
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China; Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China; Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ziqi Zhu
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Qiaoxiu Gu
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Xuqing He
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Sujuan Zhang
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Rurong Mao
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Jingliang Xu
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China; Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Fengshan Xie
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Hui Wang
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Ziteng Li
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Jing Shu
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Weifeng Zhang
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Yulin Sha
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Jin Huang
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Meng Su
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Qu Zheng
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Jian Ma
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Xiaolin Zhou
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Ming Li
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Haiying Pan
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Ya Li
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Meiling Yan
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Xueling Chen
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Mingzhou Li
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Keren Long
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Fanli Kong
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Chuang Tang
- Institute of Animal Genetics and Breeding, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Ju Huang
- First Affiliated Hospital of Shantou University, Shantou, China
| | - Chanjuan Su
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Jirui Li
- Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China
| | - Zheng Fang
- Motic China Group CO., LTD., Xiamen, China
| | | | - Erpo Tian
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Ying Zhong
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China
| | - Jiang Gu
- Jinxin Research Institute for Reproductive Medicine and Genetics, Sichuan Jinxin Xi'nan Women's and Children's Hospital, Chengdu, China; Provincial Key Laboratory of Molecular Pathology and Personalized Medicine, Center of Collaborative and Creative Center, Department of Pathology and Pathophysiology, Shantou University Medical College, Shantou, China.
| |
Collapse
|
21
|
Qin J, Lv B, Yao Y, Han X, Xue Z, Lin CP, Xue J, Ji Y. CTNND1 affects trophoblast proliferation and specification during human embryo implantation. Biol Reprod 2025; 112:46-53. [PMID: 39561119 DOI: 10.1093/biolre/ioae163] [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: 06/26/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 11/21/2024] Open
Abstract
The placenta, serving as the crucial link between maternal and infant, plays a pivotal role in maintaining a healthy pregnancy. Placental dysplasia can lead to various complications, underscoring the importance of understanding trophoblast lineage development. During peri-implantation, the trophectoderm undergoes differentiation into cytotrophoblast, syncytiotrophoblast, and extravillous trophoblast. However, the specification and regulation of human trophoblast lineage during embryo implantation, particularly in the peri-implantation phase, remain to be explored. In this study, we employed a co-culture model of human endometrial cells and native embryos and analyzed the single-cell transcriptomic data of 491 human embryonic trophoblasts during E6 to E10 to identify the key regulatory factors and the lineage differentiation process during peri-implantation. Our data identified four cell subpopulations during the implantation, including a specific transitional state toward the differentiation in which the CTNND1, one crucial component of Wnt signaling pathway activated by cadherins, acted as a crucial factor. Knockdown of CTNND1 impacted the proliferative capacity of human trophoblast stem cells, leading to early extravillous trophoblast-like differentiation. Intriguingly, ablation of CTNND1 compromised the terminal differentiation of human trophoblast stem cells toward syncytiotrophoblast or extravillous trophoblast in vitro. These findings contribute valuable insights into trophoblast lineage dynamics and offer a reference for research on placental-related diseases.
Collapse
Affiliation(s)
- Jiaying Qin
- Stem Cell Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Bo Lv
- Stem Cell Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yao Yao
- Department of Reproductive center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Xuan Han
- Stem Cell Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Zhigang Xue
- Stem Cell Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Chao-Po Lin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| | - Jinfeng Xue
- Stem Cell Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| | - Yazhong Ji
- Department of Reproductive center, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, China
| |
Collapse
|
22
|
Unjitwattana T, Huang Q, Yang Y, Tao L, Yang Y, Zhou M, Du Y, Garmire LX. Single-cell RNA-Seq data have prevalent blood contamination but can be rescued by Originator, a computational tool separating single-cell RNA-Seq by genetic and contextual information. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.04.588144. [PMID: 38617220 PMCID: PMC11014553 DOI: 10.1101/2024.04.04.588144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Single-cell RNA sequencing (scRNA-Seq) data from complex human tissues have prevalent blood cell contamination during the sample preparation process. They may also comprise cells of different genetic makeups. These issues demand rigorous preprocessing and filtering prior to the downstream functional analysis, to avoid biased conclusions due to cell types not of interest. Towards this, we propose a new computational framework, Originator, which deciphers single cells by the genetic origin and separates immune cells of blood contamination from those of expected tissue-resident cells. We demonstrate the accuracy of Originator at separating immune cells from the blood and tissue as well as cells of different genetic origins, using a variety of artificially mixed and real datasets. We show the prevalence of blood contamination in scRNA-Seq data of many tissue types. We alert the significant consequences if failing to adjust for these confounders, using scRNA-Seq data of pancreatic cancer and placentas as examples.
Collapse
Affiliation(s)
- Thatchayut Unjitwattana
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Ann Arbor, MI, 48109, USA
| | - Qianhui Huang
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave Ann Arbor, MI, 48109, USA
| | - Yiwen Yang
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave Ann Arbor, MI, 48109, USA
| | - Leyang Tao
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Ann Arbor, MI, 48109, USA
| | - Youqi Yang
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, USA
| | - Mengtian Zhou
- Department of Statistics, University of Michigan,1085 S University Ave, Ann Arbor, MI, 48109, USA
| | - Yuheng Du
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave Ann Arbor, MI, 48109, USA
| | - Lana X. Garmire
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel, Ann Arbor, MI, 48109, USA
- Department of Computation Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Ave Ann Arbor, MI, 48109, USA
| |
Collapse
|
23
|
Doratt BM, True HE, Sureshchandra S, Qiao Q, Rincon M, Marshall NE, Messaoudi I. The immune landscape of fetal chorionic villous tissue in term placenta. Front Immunol 2025; 15:1506305. [PMID: 39872537 PMCID: PMC11769816 DOI: 10.3389/fimmu.2024.1506305] [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: 10/04/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025] Open
Abstract
Introduction The immune compartment within fetal chorionic villi is comprised of fetal Hofbauer cells (HBC) and invading placenta-associated maternal monocytes and macrophages (PAMM). Recent studies have characterized the transcriptional profile of the first trimester (T1) placenta; however, the phenotypic and functional diversity of chorionic villous immune cells at term (T3) remain poorly understood. Methods To address this knowledge gap, immune cells from human chorionic villous tissues obtained from full-term, uncomplicated pregnancies were deeply phenotyped using a combination of flow cytometry, single-cell RNA sequencing (scRNA-seq, CITE-seq) and chromatin accessibility profiling (snATAC-seq). Results Our results indicate that, relative to the first trimester, the frequency of fetal macrophages (HBC, proliferating HBC) is significantly reduced, whereas that of infiltrating maternal monocytes/macrophages (PAMM1b, PAMM1a, PAMM2, MAC_1) increased in T3. PAMM1b and HBCs exhibit the most phagocytic capacity at term highlighting their regulatory role in tissue homeostasis in late pregnancy. The transcriptional profiles of resident villous immune subsets exhibit a heightened activation state relative to the relative to T1, likely to support labor and parturition. Additionally, we provide one of the first insights into the chromatin accessibility profile of villous myeloid cells at term. We next stratified our findings by pre-pregnancy BMI since maternal pregravid obesity is associated with several adverse pregnancy outcomes. Pregravid obesity increased inflammatory gene expression, particularly among HBC and PAMM1a subsets, but dampened the expression of antimicrobial genes, supporting a tolerant-like phenotype of chorionic villous myeloid cells. We report a decline in HBC abundance accompanied by an increase in infiltrating maternal macrophages, which aligns with reports of heightened chorionic villous inflammatory pathologies with pregravid obesity. Finally, given the shared fetal yolk-sac origin of HBCs and microglia, we leveraged an in vitro model of umbilical cord blood-derived microglia to investigate the impact of pregravid obesity on fetal neurodevelopment. Our findings reveal increased expression of activation markers albeit dampened phagocytic capacity in microglia with pregravid obesity. Discussion Overall, our study highlights immune adaptations in the fetal chorionic villous with gestational age and pregravid obesity, as well as insight towards microglia dysfunction possibly underlying poor neurodevelopmental outcomes in offspring of women with pregravid obesity.
Collapse
Affiliation(s)
- Brianna M. Doratt
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| | - Heather E. True
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, United States
| | - Suhas Sureshchandra
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA, United States
- Institute for Immunology, University of California, Irvine, Irvine, CA, United States
| | - Qi Qiao
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
- Department of Biostatistics, University of Kentucky, Lexington, KY, United States
| | - Monica Rincon
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Nicole E. Marshall
- Maternal-Fetal Medicine, Oregon Health and Science University, Portland, OR, United States
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
24
|
Uhm C, Gu J, Ju W, Pizzella S, Oktay H, Peng JYC, Guariglia S, Liu Y, Zhao H, Wang Y, Menon R, Zhong N. Single-nucleus RNA sequencing reveals distinct pathophysiological trophoblast signatures in spontaneous preterm birth subtypes. Cell Biosci 2025; 15:1. [PMID: 39773245 PMCID: PMC11705668 DOI: 10.1186/s13578-024-01343-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Spontaneous preterm birth (sPTB) poses significant challenges, affecting neonatal health and neurodevelopmental outcomes worldwide. The specific effects of placental trophoblasts on the pathological development of sPTB subtypes-preterm premature rupture of fetal membranes (pPROM) and spontaneous preterm labor (sPTL)-are not fully understood, making it crucial to uncover these impacts for the development of effective therapeutic strategies. Using single-nucleus RNA sequencing, we investigated transcriptomic and cellular differences at the maternal-fetal interface in pPROM and sPTL placentas. Our findings revealed distinct trophoblast compositions with pPROM characterized predominantly by extravillous trophoblasts (EVTs), while sPTL showed an abundance of syncytiotrophoblasts (STBs). Through cell differentiation and cell-to-cell communication analyses, other distinguishing factors were also found. In pPROM, heightened inflammation, oxidative stress, and vascular dysregulation with key pathways including tumor necrosis factor signaling, matrix metalloproteinase activation, and integrin-mediated cell adhesion, highlighted an invasive EVT profile potentially driven by hypoxic conditions and immune cell recruitment. In contrast, sPTL was marked by increased smooth muscle contraction, vascular remodeling, and altered signaling dynamics involving fibroblasts, including TGF-β and WNT pathways. Our study highlights the critical need to distinguish sPTB subtypes to improve diagnostic precision and therapeutic targeting. The molecular insights gained provide a foundation for future investigations aimed at functional validation of key pathways and exploration of trophoblasts on the development of sPTB. Ultimately, these findings pave the way for more personalized and effective interventions to mitigate adverse outcomes associated with preterm birth.
Collapse
Affiliation(s)
- Cherilyn Uhm
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Jianlei Gu
- School of Public Health, Yale University, New Haven, CT, 06520-0834, USA
| | - Weina Ju
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Stephanie Pizzella
- Department of Obstetrics and Gynecology, School of Medicine, Washington University, St. Louis, MO, 63110-1010, USA
| | - Hande Oktay
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | | | - Sararose Guariglia
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Yong Liu
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Hongyu Zhao
- School of Public Health, Yale University, New Haven, CT, 06520-0834, USA
| | - Yong Wang
- Department of Obstetrics and Gynecology, School of Medicine, Washington University, St. Louis, MO, 63110-1010, USA
| | - Ramkumar Menon
- The University of Texas Medical Branch at Galveston, Galveston, TX, 77555-0144, USA
| | - Nanbert Zhong
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA.
| |
Collapse
|
25
|
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
|
26
|
Chen X, Tober J, Dominguez M, Tang AT, Bockman J, Yang J, Mani S, Lee CN, Chen M, Thillaikumaran T, Mericko-Ishizuka P, Mainigi M, Speck NA, Kahn ML. Lineage tracing studies suggest that the placenta is not a de novo source of hematopoietic stem cells. PLoS Biol 2025; 23:e3003003. [PMID: 39874373 PMCID: PMC11774391 DOI: 10.1371/journal.pbio.3003003] [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: 08/14/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025] Open
Abstract
Definitive hematopoietic stem and progenitor cells (HSPCs) arise from a small number of hemogenic endothelial cells (HECs) within the developing embryo. Understanding the origin and ontogeny of HSPCs is of considerable interest and potential therapeutic value. It has been proposed that the murine placenta contains HECs that differentiate into HSPCs. However, during human gestation HSPCs arise in the aorta considerably earlier than when they can first be detected in the placenta, suggesting that the placenta may primarily serve as a niche. We found that the Runx1 transcription factor, which is required to generate HSPCs from HECs, is not expressed by mouse placental ECs. To definitively determine whether the mouse placenta is a site of HSPC emergence, we performed lineage tracing experiments with a Hoxa13Cre allele that specifically labels ECs in the placenta and umbilical cord (UC), but not in the yolk sac or embryo. Immunostaining revealed Hoxa13Cre lineage-traced HECs and HSPCs in the UC, a known site of HECs, but not the placenta. Consistent with these findings, ECs harvested from the E10.5 aorta and UC, but not the placenta, gave rise to hematopoietic cells ex vivo, while colony forming assays using E14.5 fetal liver revealed only 2% of HSPCs arose from Hoxa13-expressing precursors. In contrast, the pan-EC Cdh5-CreERT2 allele labeled most HSPCs in the mouse placenta. Lastly, we found that RUNX1 and other HEC genes were not expressed in first-trimester human placenta villous ECs, suggesting that human placenta is not hemogenic. Our findings demonstrate that the placenta functions as a site for expansion of HSPCs that arise within the embryo proper and is not a primary site of HSPC emergence.
Collapse
Affiliation(s)
- Xiaowen Chen
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Joanna Tober
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Martin Dominguez
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Alan T. Tang
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jenna Bockman
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jisheng Yang
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Sneha Mani
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Chin Nien Lee
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mei Chen
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Triloshan Thillaikumaran
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Patricia Mericko-Ishizuka
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Monica Mainigi
- Center for Research on Reproduction and Women’s Health, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nancy A. Speck
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Mark L. Kahn
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
27
|
Qin X, Zhang M, Chen S, Tang Y, Cui J, Ding G. Short-chain fatty acids in fetal development and metabolism. Trends Mol Med 2024:S1471-4914(24)00329-0. [PMID: 39694776 DOI: 10.1016/j.molmed.2024.11.014] [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: 08/31/2024] [Revised: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024]
Abstract
Short-chain fatty acids (SCFAs), primarily derived from gut microbiota, play a role in regulating fetal development; however, the mechanism remains unclear. Fetal SCFAs levels depends on maternal SCFAs transported via the placenta. Metabolic stress, particularly from diabetes and obesity, can disrupt maternal SCFAs levels, impairing fetal metabolic reprogramming. Dysregulated SCFAs may negatively impact the development of the fetal cardiovascular, nervous, and immune systems, potentially contributing to adverse outcomes in adulthood. This review focuses on recent advances regarding the role of maternal SCFAs in shaping the metabolic profile of offspring, especially in the context of various maternal metabolic disorders. Given that SCFAs may influence fetal development through the placenta-embryo axis, targeted SCFAs supplementation could be a promising strategy against developmental diseases associated with intrauterine risk factors.
Collapse
Affiliation(s)
- Xueyun Qin
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Mo Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Shiting Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Yunhui Tang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Jiajun Cui
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China; Shanghai Key Laboratory of Reproduction and Development, Shanghai, China; Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
28
|
Matsumoto S, Tanaka S. Wnt signaling activation confers a syncytiotrophoblast progenitor state on trophoblast stem cells of cynomolgus monkey†. Biol Reprod 2024; 111:1262-1281. [PMID: 39223948 PMCID: PMC11647102 DOI: 10.1093/biolre/ioae131] [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: 02/29/2024] [Revised: 06/08/2024] [Accepted: 09/03/2024] [Indexed: 09/04/2024] Open
Abstract
Trophoblast stem cells, derived from the trophectoderm of the blastocyst, are used as an in vitro model to reveal the mechanisms underlying placentation in mammals. In humans, suitable culture conditions for trophoblast stem cell derivation have recently been established. The established human trophoblast stem cells differentiate efficiently toward two trophoblast subtypes: syncytiotrophoblasts and extravillous trophoblasts. However, the efficiency of differentiation is lower in macaque trophoblast stem cells than in human trophoblast stem cells. Here, we demonstrate that the activation of Wnt signaling downregulated the expression of inhibitory G protein and induced trophoblastic lineage switching to the syncytiotrophoblast progenitor state. The treatment of macaque trophoblast stem cells with a GSK-3 inhibitor, CHIR99021, upregulated syncytiotrophoblast progenitor markers and enhanced proliferation. Under the Wnt signaling-activated conditions, macaque trophoblast stem cells effectively differentiated to syncytiotrophoblasts upon dibutyryl cyclic AMP (dbcAMP) and forskolin treatment. RNA-seq analyses revealed the downregulation of inhibitory G protein, which may make macaque trophoblast stem cells responsive to forskolin. Interestingly, this lineage switching appeared to be reversible as the macaque trophoblast stem cells lost responsiveness to forskolin upon the removal of CHIR99021. The ability to regulate the direction of macaque trophoblast stem cell differentiation would be advantageous in elucidating the mechanisms underlying placentation in non-human primates.
Collapse
Affiliation(s)
- Shoma Matsumoto
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Satoshi Tanaka
- Department of Animal Resource Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
29
|
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
|
30
|
Liu X, Xin S, Xu F, Zhou M, Xiong Y, Zeng Y, Zeng X, Zou Y. Single-cell RNA sequencing reveals heterogeneity and differential expression of the maternal-fetal interface during ICP and normal pregnancy. J Matern Fetal Neonatal Med 2024; 37:2361278. [PMID: 38835155 DOI: 10.1080/14767058.2024.2361278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 05/24/2024] [Indexed: 06/06/2024]
Abstract
OBJECTIVE Intrahepatic cholestasis of pregnancy (ICP) can cause adverse perinatal outcomes. Previous studies have demonstrated that the placenta of an ICP pregnancy differs in morphology and gene expression from the placenta of a normal pregnancy. To date, however, the genetic mechanism by which ICP affects the placenta is poorly understood. Therefore, the aim of this study was to investigate the differences in main cell types, gene signatures, cell ratio, and functional changes in the placenta between ICP and normal pregnancy. METHODS Single-cell RNA sequencing (scRNA-seq) technology was used to detect the gene expression of all cells at the placental maternal-fetal interface. Two individuals were analyzed - one with ICP and one without ICP. The classification of cell types was determined by a graph-based clustering algorithm. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using the R software phyper () function and DAVID website. The differentially expressed genes (DEGs) encoding transcription factors (TFs) were identified using getorf and DIAMOND software. RESULTS We identified 14 cell types and 22 distinct cell subtypes that showed unique functional properties. Additionally, we found differences in the proportions of fibroblasts 1, helper T (Th) cells, extravillous trophoblasts, and villous cytotrophoblasts, and we observed heterogeneity of gene expression between ICP and control placentas. Furthermore, we identified 263 DEGs that belonged to TF families, including zf-C2H2, HMGI/HMGY, and Homeobox. In addition, 28 imprinted genes were preferentially expressed in specific cell types, such as PEG3 and PEG10 in trophoblasts as well as DLK1 and DIO3 in fibroblasts. CONCLUSIONS Our results revealed the differences in cell-type ratios, gene expression, and functional changes between ICP and normal placentas, and heterogeneity was found among cell subgroups. Hence, the imbalance of various cell types affects placental activity to varying degrees, indicating the complexity of the cell networks that form the placental tissue system, and this alteration of placental function is associated with adverse events in the perinatal period.
Collapse
Affiliation(s)
- Xianxian Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Siming Xin
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Fangping Xu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Mengni Zhou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Ying Xiong
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Yang Zeng
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Xiaoming Zeng
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| | - Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi, China
| |
Collapse
|
31
|
Rong L, Xiang L, Ai Z, Niu B, Wang Y, Yin Y, Feng C, Shi G, Chen T, Yang J, Luo X, Bai Y, Zhou X, Liu X, Zheng H, Ke Y, Li T, Wu Z. The impact of dynamic caudal type homeobox 2 expression on the differentiation of human trophoblast lineage during implantation. Cell Prolif 2024; 57:e13729. [PMID: 39161168 PMCID: PMC11628739 DOI: 10.1111/cpr.13729] [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: 04/30/2024] [Revised: 06/21/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024] Open
Abstract
The trophoblast lineage differentiation represents a rate-limiting step in successful embryo implantation. Adhesion, invasion and migration processes within the trophoblast are governed by several transcription factors. Among them, CDX2 is a critical regulator shaping the destiny of the trophoblast. While its altered expression is a linchpin initiating embryo implantation in mice, the precise influence of CDX2 on the functionality and lineage differentiation of early human trophoblast remains unclear. In this study, we employed well-established human trophoblast stem cell (hTSC) lines with CDX2 overexpression coupled with a 3D in vitro culture system for early human embryos. We revealed that the downregulation of CDX2 is a prerequisite for syncytialization during human embryo implantation based on immunofluorescence, transcriptome analysis, CUT-tag sequencing and the construction of 3D human trophoblast organoids. While CDX2 overexpression inhibited syncytialization, it propelled hTSC proliferation and invasive migration. CDX2 exerted its influence by interacting with CGA, PTGS2, GCM1, LEF1 and CDH2, thereby hindering premature differentiation of the syncytiotrophoblast. CDX2 overexpression enhanced the epithelial-mesenchymal transition of human trophoblast organoids. In summary, our study provides insights into the molecular characteristics of trophoblast differentiation and development in humans, laying a theoretical foundation for advancing research in embryo implantation.
Collapse
Affiliation(s)
- Lujuan Rong
- Faculty of Life Science and TechnologyKunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Lifeng Xiang
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Zongyong Ai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Baohua Niu
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Yaqing Wang
- University of Science and Technology of ChinaHefeiAnhuiChina
- Suzhou Institute for Advanced ResearchUniversity of Science and Technology of ChinaSuzhouJiangsuChina
| | - Yu Yin
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Chun Feng
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Gaohui Shi
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Tingwei Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Jie Yang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Xi Luo
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Yun Bai
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Xiaoting Zhou
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Xiaoping Liu
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Haishan Zheng
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
| | - Yang Ke
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnanChina
| | - Ze Wu
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- The Affiliated Hospital of Kunming University of Science and TechnologyKunmingYunnanChina
- Department of Reproductive Medicine, NHC Key Laboratory of Healthy Birth and Birth Defect Prevention in Western China (Co‐building)The First People's Hospital of Yunnan ProvinceKunmingYunnanChina
- KUST‐YPFPH Reproductive Medicine Joint Research CenterKunmingYunnanChina
| |
Collapse
|
32
|
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
|
33
|
Zhang Y, Yang L, Yang D, Cai S, Wang Y, Wang L, Li Y, Li L, Yin T, Diao L. Understanding the heterogeneity of natural killer cells at the maternal-fetal interface: implications for pregnancy health and disease. Mol Hum Reprod 2024; 30:gaae040. [PMID: 39570646 DOI: 10.1093/molehr/gaae040] [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: 07/10/2024] [Revised: 11/09/2024] [Indexed: 11/22/2024] Open
Abstract
Natural killer (NK) cells are the most abundant leukocytes located at the maternal-fetal interface; they respond to pregnancy-related hormones and play a pivotal role in maintaining the homeostatic micro-environment during pregnancy. However, due to the high heterogeneity of NK cell subsets, their categorization has been controversial. Here, we review previous studies on uterine NK cell subsets, including the classic categorization based on surface markers, functional molecules, and developmental stages, as well as single-cell RNA sequencing-based clustering approaches. In addition, we summarize the potential pathways by which endometrial NK cells differentiate into decidual NK (dNK) cells, as well as the differentiation pathways of various dNK subsets. Finally, we compared the alterations in the NK cell subsets in various pregnancy-associated diseases, emphasizing the possible contribution of specific subsets to the development of the disease.
Collapse
Affiliation(s)
- Yuying Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Liangtao Yang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, China
| | - Dongyong Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Songchen Cai
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, China
| | - Yanjun Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Linlin Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, China
| | - Yuye Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, China
| | - Longfei Li
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, China
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproductive Medicine and Genetics, Shenzhen Zhongshan Obstetrics & Gynecology Hospital (formerly Shenzhen Zhongshan Urology Hospital), Shenzhen, China
- Guangdong Engineering Technology Research Center of Reproductive Immunology for Peri-Implantation, Shenzhen, China
| |
Collapse
|
34
|
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
|
35
|
Slamecka J, Ryu S, Tristan CA, Chu PH, Weber C, Deng T, Gedik Y, Ormanoglu P, Voss TC, Simeonov A, Singeç I. Highly efficient generation of self-renewing trophoblast from human pluripotent stem cells. iScience 2024; 27:110874. [PMID: 39386760 PMCID: PMC11462042 DOI: 10.1016/j.isci.2024.110874] [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: 12/18/2023] [Revised: 06/02/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Human pluripotent stem cells (hPSCs) represent a powerful model system to study early developmental processes. However, lineage specification into trophectoderm (TE) and trophoblast (TB) differentiation remains poorly understood, and access to well-characterized placental cells for biomedical research is limited, largely depending on fetal tissues or cancer cell lines. Here, we developed novel strategies enabling highly efficient TE specification that generates cytotrophoblast (CTB) and multinucleated syncytiotrophoblast (STB), followed by the establishment of trophoblast stem cells (TSCs) capable of differentiating into extravillous trophoblast (EVT) and STB after long-term expansion. We confirmed stepwise and controlled induction of lineage- and cell-type-specific genes consistent with developmental biology principles and benchmarked typical features of placental cells using morphological, biochemical, genomics, epigenomics, and single-cell analyses. Charting a well-defined roadmap from hPSCs to distinct placental phenotypes provides invaluable opportunities for studying early human development, infertility, and pregnancy-associated diseases.
Collapse
Affiliation(s)
- Jaroslav Slamecka
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Seungmi Ryu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Carlos A. Tristan
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pei-Hsuan Chu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Claire Weber
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Tao Deng
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Yeliz Gedik
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Pinar Ormanoglu
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ty C. Voss
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| | - Ilyas Singeç
- National Center for Advancing Translational Sciences (NCATS), Division of Preclinical Innovation, Stem Cell Translation Laboratory (SCTL), National Institutes of Health (NIH), Rockville, MD 20850, USA
| |
Collapse
|
36
|
Kalisch-Smith JI, Ehtisham-Uddin N, Rodriguez-Caro H. Feto-placental and coronary endothelial genes implicated in miscarriage, congenital heart disease and stillbirth, a systematic review and meta-analysis. Placenta 2024; 156:55-66. [PMID: 39276426 DOI: 10.1016/j.placenta.2024.08.015] [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: 07/08/2024] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024]
Abstract
The first trimester placenta is very rarely investigated for placental vascular formation in developmental or diseased contexts. Defects in placental formation can cause heart defects in the fetus, and vice versa. Determining the causality is therefore difficult as both organs develop concurrently and express many of the same genes. Here, we performed a systematic review to determine feto-placental and coronary endothelial genes implicated in miscarriages, stillbirth and congenital heart defects (CHD) from human genome wide screening studies. 4 single cell RNAseq datasets from human first/early second trimester cardiac and placental samples were queried to generate a list of 1187 endothelial genes. This broad list was cross-referenced with genes implicated in the pregnancy disorders above. 39 papers reported feto-placental and cardiac coronary endothelial genes, totalling 612 variants. Vascular gene variants were attributed to the incidence of miscarriage (8 %), CHD (4 %) and stillbirth (3 %). The most common genes for CHD (NOTCH, DST, FBN1, JAG1, CHD4), miscarriage (COL1A1, HERC1), and stillbirth (AKAP9, MYLK), were involved in blood vessel and cardiac valve formation, with roles in endothelial differentiation, angiogenesis, extracellular matrix signaling, growth factor binding and cell adhesion. NOTCH1, AKAP12, CHD4, LAMC1 and SOS1 showed greater relative risk ratios with CHD. Many of the vascular genes identified were expressed highly in both placental and heart EC populations. Both feto-placental and cardiac vascular genes are likely to result in poor endothelial cell development and function during human pregnancy that leads to higher risk of miscarriage, congenital heart disease and stillbirth.
Collapse
Affiliation(s)
- Jacinta I Kalisch-Smith
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX3 7TY, UK.
| | - Nusaybah Ehtisham-Uddin
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX3 7TY, UK
| | - Helena Rodriguez-Caro
- Institute for Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX3 7TY, UK; Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research, Lausanne, Switzerland
| |
Collapse
|
37
|
Chen EX, Hu SC, Xu JQ, Liu KY, Tang J, Shen XP, Liang X, Xie YL, Ge LX, Luo X, Wang YX, Xiang YL, Ding YB. Suppression of GATA3 promotes epithelial-mesenchymal transition and simultaneous cellular senescence in human extravillous trophoblasts. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119768. [PMID: 38838858 DOI: 10.1016/j.bbamcr.2024.119768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 06/07/2024]
Abstract
The regulatory mechanism of the transcription factor GATA3 in the differentiation and maturation process of extravillous trophoblasts (EVT) in early pregnancy placenta, as well as its relevance to the occurrence of pregnancy disorders, remains poorly understood. This study leveraged single-cell RNA sequencing data from placental organoid models and placental tissue to explore the dynamic changes in GATA3 expression during EVT maturation. The expression pattern exhibited an initial upregulation followed by subsequent downregulation, with aberrant GATA3 localization observed in cases of recurrent miscarriage (RM). By identifying global targets regulated by GATA3 in primary placental EVT cells, JEG3, and HTR8/SVneo cell lines, this study offered insights into its regulatory mechanisms across different EVT cell models. Shared regulatory targets among these cell types and activation of trophoblast cell marker genes emphasized the importance of GATA3 in EVT differentiation and maturation. Knockdown of GATA3 in JEG3 cells led to repression of GATA3-induced epithelial-mesenchymal transition (EMT), as evidenced by changes in marker gene expression levels and enhanced migration ability. Additionally, interference with GATA3 accelerated cellular senescence, as indicated by reduced proliferation rates and increased activity levels for senescence-associated β-galactosidase enzyme, along with elevated expression levels for senescence-associated genes. This study provides comprehensive insights into the dual role of GATA3 in regulating EMT and cellular senescence during EVT differentiation, shedding light on the dynamic changes in GATA3 expression in normal and pathological placental conditions.
Collapse
Affiliation(s)
- En-Xiang Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China; Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Department of Basic Medical Sciences, Changsha Medical University, Hunan 410219, China
| | - Si-Chen Hu
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Jia-Qi Xu
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Kun-Yan Liu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jing Tang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China; Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Xi-Peng Shen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Xiao Liang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - You-Long Xie
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Lu-Xin Ge
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China; Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University. Hunan 410219, China
| | - Xin Luo
- Department of Obstetrics, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying-Xiong Wang
- Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| | - Yun-Long Xiang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China.
| | - Yu-Bin Ding
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing 401147, China; Department of Toxicology, Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
38
|
Vachalova V, Kumnova F, Synova T, Anandam KY, Abad C, Karahoda R, Staud F. Metformin inhibits OCT3-mediated serotonin transport in the placenta. Biomed Pharmacother 2024; 179:117399. [PMID: 39243433 DOI: 10.1016/j.biopha.2024.117399] [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: 06/04/2024] [Revised: 08/19/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024] Open
Abstract
Proper fetal development requires tight regulation of serotonin concentrations within the fetoplacental unit. This homeostasis is partly maintained by the placental transporter OCT3/SLC22A3, which takes up serotonin from the fetal circulation. Metformin, an antidiabetic drug commonly used to treat gestational diabetes mellitus, was shown to inhibit OCT3. We, therefore, hypothesized that its use during pregnancy could disrupt placental serotonin homeostasis. This hypothesis was tested using three experimental model systems: primary trophoblast cells isolated from the human term placenta, fresh villous human term placenta fragments, and rat term placenta perfusions. Inhibition of serotonin transport by metformin at three concentrations (1 μM, 10 μM, and 100 μM) was assessed in all three models. The OCT3 inhibitor decynium-22 (100 μM) and paroxetine (100 μM), a dual inhibitor of SERT and OCT3, were used as controls. In primary trophoblasts, paroxetine exhibited the strongest inhibition of serotonin uptake, followed by decynium-22. Metformin showed a concentration-dependent effect, reducing serotonin uptake by up to 57 % at the highest concentration. Its inhibitory effect was less pronounced in fresh villous fragments but remained statistically significant at all concentrations. In the perfused rat placenta, metformin demonstrated a concentration-dependent effect, reducing placental serotonin uptake by 44 % at the highest concentration tested. Our findings across all experimental models show inhibition of placental OCT3 by metformin, resulting in reduced serotonin uptake by the trophoblast. This sheds light on mechanisms that may underpin metformin-mediated effects on fetal development.
Collapse
Affiliation(s)
- Veronika Vachalova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Fiona Kumnova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Tetiana Synova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Kasin Yadunandam Anandam
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Cilia Abad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Rona Karahoda
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic
| | - Frantisek Staud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Hradec Kralove, Czech Republic.
| |
Collapse
|
39
|
Tang M, Xiong L, Cai J, Gong X, Fan L, Zhou X, Xing S, Yang X. Comprehensive Analysis of scRNA-Seq and Bulk RNA-Seq Reveals Transcriptional Signatures of Macrophages in Intrahepatic Cholestasis of Pregnancy. J Inflamm Res 2024; 17:6863-6874. [PMID: 39372590 PMCID: PMC11451404 DOI: 10.2147/jir.s471374] [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: 05/29/2024] [Accepted: 09/26/2024] [Indexed: 10/08/2024] Open
Abstract
Purpose Intrahepatic cholestasis of pregnancy (ICP) is a disorder that characterized by maternal pruritus, abnormal liver function, and an elevation in total bile acid concentrations during pregnancy. Immune factors have been recognized as playing a vital role in the mechanism of ICP. However, the underlying mechanisms regulating dysfunctional immune cells and immune genes remain to be fully elucidated. Patients and Methods Single-cell RNA sequencing and bulk RNA sequencing data of the placenta were downloaded from the SRA database. The AUCell package, Monocle package and SCENIC package were utilized to explored immune cell activity, cell trajectory and transcription factor, respectively. GO, KEGG, and GSEA were employed to explore potential biological mechanisms. Cell-cell communications were further investigated using the CellChat package. RT-PCR, and Western blot were used to verify the gene expression in placenta. Results In placenta cells, macrophages were found to be significantly increased in ICP. Additionally, macrophages exhibited the highest immune gene score and were divided into four subclusters (MF1-4). Our analysis revealed significant elevations in MF2, associated with LPS response and antigen presentation, and MF4, associated with TNF and cytokine production. MF3 displayed an anti-inflammatory phenotype. MF1, closely related to ribosomes and proteins, exhibited a sharp decrease. Although ICP maintained an anti-inflammatory state, macrophage trajectories showed a gradual progression toward inflammation. Subsequently, we confirmed that cytokine- and chemokine-related signaling pathways were emphasized in macrophages. Within the CXCL signaling pathway, the increased expression of CXCL1 in macrophages can interact with CXCR2 in neutrophils, potentially inducing macrophage infiltration, stimulating neutrophil chemotaxis, and leading to an inflammatory response and cellular damage. Conclusion In conclusion, we firstly revealed the transcriptional signatures of macrophages in ICP and discovered a tendency toward an inflammatory state. This study also provides new evidence that the CXCL1-CXCR2 axis may play an important role in the pathogenesis of ICP.
Collapse
Affiliation(s)
- Mi Tang
- Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, People’s Republic of China
- School of medicine, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Liling Xiong
- Obstetrics Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, People’s Republic of China
| | - Jianghui Cai
- School of medicine, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
- Department of Pharmacy, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, People’s Republic of China
| | - Xuejia Gong
- School of medicine, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Li Fan
- Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, People’s Republic of China
| | - Xiaoyu Zhou
- Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, People’s Republic of China
| | - Shasha Xing
- Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, People’s Republic of China
| | - Xiao Yang
- Obstetrics Department, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, People’s Republic of China
| |
Collapse
|
40
|
Stadtmauer DJ, Basanta Martínez S, Maziarz JD, Cole AG, Dagdas G, Smith GR, van Breukelen F, Pavličev M, Wagner GP. Cell type and cell signaling innovations underlying mammalian pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.591945. [PMID: 38746137 PMCID: PMC11092578 DOI: 10.1101/2024.05.01.591945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
How fetal and maternal cell types have co-evolved to enable mammalian placentation poses a unique evolutionary puzzle. Here, we present a multi-species atlas integrating single-cell transcriptomes from six species bracketing therian mammal diversity. We find that invasive trophoblasts share a gene-expression signature across eutherians, and evidence that endocrine decidual cells evolved stepwise from an immunomodulatory cell type retained in Tenrec with affinity to human decidua of menstruation. We recover evolutionary patterns in ligand-receptor signaling: fetal and maternal cells show a pronounced tendency towards disambiguation, but a predicted arms race dynamic between them is limited. We reconstruct cell communication networks of extinct mammalian ancestors, finding strong integration of fetal trophoblast into maternal networks. Together, our results reveal a dynamic history of cell type and signaling evolution. Synopsis The fetal-maternal interface is one of the most intense loci of cell-cell signaling in the human body. Invasion of cells from the fetal placenta into the uterus, and the corresponding transformation of maternal tissues called decidualization, first evolved in the stem lineage of eutherian mammals( 1 , 2 ). Single-cell studies of the human fetal-maternal interface have provided new insight into the cell type diversity and cell-cell interactions governing this chimeric organ( 3-5 ). However, the fetal-maternal interface is also one of the most rapidly evolving, and hence most diverse, characters among mammals( 6 ), and an evolutionary analysis is missing. Here, we present and compare single-cell data from the fetal-maternal interface of species bracketing key events in mammal phylogeny: a marsupial (opossum, Monodelphis domestica ), the afrotherian Tenrec ecaudatus, and four Euarchontoglires - guinea pig and mouse (Rodentia) together with recent macaque and human data (primates) ( 4 , 5 , 7 ). We infer cell type homologies, identify a gene-expression signature of eutherian invasive trophoblast conserved over 99 million years, and discover a predecidual cell in the tenrec which suggests stepwise evolution of the decidual stromal cell. We reconstruct ancestral cell signaling networks, revealing the integration of fetal cell types into the interface. Finally, we test two long-standing theoretical predictions, the disambiguation hypothesis( 8 ) and escalation hypothesis( 9 ), at transcriptome-wide scale, finding divergence between fetal and maternal signaling repertoires but arms race dynamics restricted to a small subset of ligand-receptor pairs. In so doing, we trace the co-evolutionary history of cell types and their signaling across mammalian viviparity.
Collapse
|
41
|
Liu Z. Gene expression profile of human placental villous pericytes in the first trimester - An analysis by single-cell RNA sequencing. Reprod Biol 2024; 24:100919. [PMID: 38941941 DOI: 10.1016/j.repbio.2024.100919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024]
Abstract
Mesenchymal cells within theplacental villi play a crucial role in shaping the morphology of branching structures and driving the development of blood vessels. However, the markers and functions of placental villous pericytes (PVPs) as distinct subgroups of placental villous mesenchymal cells, remain unclear. Therefore, in this study, the markers and functions of PVPs were investigated. Single-cell sequencing data from the first-trimester placental villi was obtained and the Seurat tool was used to identify PVP markers. Gene Ontology (GO) analysis of specific genes was performed using the DAVID database. The Cellchat tool was employed to investigate the interaction signals between the PVPs and other cells. Expression of the PVP markers was confirmed using immunofluorescence. Presence of extracellular vesicles in the placental villous mesenchyme and PVPs was examined by transmission electron microscopy. Our findings revealed that renin (REN) and amphiregulin (AREG)-positive fibroblasts in the placental villi specifically expressed several classic pericyte markers. In the first trimester, certain conserved functions of pericytes were observed and they displayed tissue-specific functions such as in the integrin-mediated signaling pathway and extracellular exosomes. Moreover, the placental villous mesenchyme was found to be rich in extracellular vesicles. AREG is specifically transcribed in the first trimester PVPs, however, its protein was located in syncytiotrophoblasts. These insights contribute to a comprehensive understanding of early placental development and offer new therapeutic targets for placenta-derived pregnancy complications.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
42
|
Yu J, Yan Y, Li S, Xu Y, Parolia A, Rizvi S, Wang W, Zhai Y, Xiao R, Li X, Liao P, Zhou J, Okla K, Lin H, Lin X, Grove S, Wei S, Vatan L, Hu J, Szumilo J, Kotarski J, Freeman ZT, Skala S, Wicha M, Cho KR, Chinnaiyan AM, Schon S, Wen F, Kryczek I, Wang S, Chen L, Zou W. Progestogen-driven B7-H4 contributes to onco-fetal immune tolerance. Cell 2024; 187:4713-4732.e19. [PMID: 38968937 PMCID: PMC11344674 DOI: 10.1016/j.cell.2024.06.012] [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: 07/19/2023] [Revised: 02/09/2024] [Accepted: 06/09/2024] [Indexed: 07/07/2024]
Abstract
Immune tolerance mechanisms are shared in cancer and pregnancy. Through cross-analyzing single-cell RNA-sequencing data from multiple human cancer types and the maternal-fetal interface, we found B7-H4 (VTCN1) is an onco-fetal immune tolerance checkpoint. We showed that genetic deficiency of B7-H4 resulted in immune activation and fetal resorption in allogeneic pregnancy models. Analogously, B7-H4 contributed to MPA/DMBA-induced breast cancer progression, accompanied by CD8+ T cell exhaustion. Female hormone screening revealed that progesterone stimulated B7-H4 expression in placental and breast cancer cells. Mechanistically, progesterone receptor (PR) bound to a newly identified -58 kb enhancer, thereby mediating B7-H4 transcription via the PR-P300-BRD4 axis. PR antagonist or BRD4 degrader potentiated immunotherapy in a murine B7-H4+ breast cancer model. Thus, our work unravels a mechanistic and biological connection of a female sex hormone (progesterone) to onco-fetal immune tolerance via B7-H4 and suggests that the PR-P300-BRD4 axis is targetable for treating B7-H4+ cancer.
Collapse
Affiliation(s)
- Jiali Yu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Yijian Yan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shasha Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Ying Xu
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Abhijit Parolia
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Syed Rizvi
- Department of Chemical Engineering, University of Michigan School of Engineering, Ann Arbor, MI, USA
| | - Weichao Wang
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Yiwen Zhai
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rongxin Xiao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Xiong Li
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Peng Liao
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiajia Zhou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Karolina Okla
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Heng Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Xun Lin
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Sara Grove
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shuang Wei
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Linda Vatan
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Jiantao Hu
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Justyna Szumilo
- Department of Clinical Pathomorphology, Medical University of Lublin, Lublin, Poland
| | - Jan Kotarski
- Department of Oncological Gynecology and Gynecology, Medical University of Lublin, Lublin, Poland
| | - Zachary T Freeman
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Stephanie Skala
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Max Wicha
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Kathleen R Cho
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Arul M Chinnaiyan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Samantha Schon
- Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Fei Wen
- Department of Chemical Engineering, University of Michigan School of Engineering, Ann Arbor, MI, USA
| | - Ilona Kryczek
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA
| | - Shaomeng Wang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Weiping Zou
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA; Graduate Program in Cancer Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
43
|
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
|
44
|
Zang X, Gu S, Wang W, Shi J, Gan J, Hu Q, Zhou C, Ding Y, He Y, Jiang L, Gu T, Xu Z, Huang S, Yang H, Meng F, Li Z, Cai G, Hong L, Wu Z. Dynamic intrauterine crosstalk promotes porcine embryo implantation during early pregnancy. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1676-1696. [PMID: 38748354 DOI: 10.1007/s11427-023-2557-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/21/2024] [Indexed: 08/09/2024]
Abstract
Dynamic crosstalk between the embryo and mother is crucial during implantation. Here, we comprehensively profile the single-cell transcriptome of pig peri-implantation embryos and corresponding maternal endometrium, identifying 4 different lineages in embryos and 13 cell types in the endometrium. Cell-specific gene expression characterizes 4 distinct trophectoderm subpopulations, showing development from undifferentiated trophectoderm to polar and mural trophectoderm. Dynamic expression of genes in different types of endometrial cells illustrates their molecular response to embryos during implantation. Then, we developed a novel tool, ExtraCellTalk, generating an overall dynamic map of maternal-foetal crosstalk using uterine luminal proteins as bridges. Through cross-species comparisons, we identified a conserved RBP4/STRA6 pathway in which embryonic-derived RBP4 could target the STRA6 receptor on stromal cells to regulate the interaction with other endometrial cells. These results provide insight into the maternal-foetal crosstalk during embryo implantation and represent a valuable resource for further studies to improve embryo implantation.
Collapse
Affiliation(s)
- Xupeng Zang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
| | - Shengchen Gu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
| | - Wenjing Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
| | - Junsong Shi
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu, 527300, China
| | - Jianyu Gan
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
| | - Qun Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
| | - Chen Zhou
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
| | - Yue Ding
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
| | - Yanjuan He
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
| | - Lei Jiang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Ting Gu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou, 510520, China
| | - Zheng Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou, 510520, China
| | - Sixiu Huang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou, 510520, China
| | - Huaqiang Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou, 510520, China
| | - Fanming Meng
- Guangdong Key Laboratory of Animal Breeding and Nutrition, Institute of Animal Science, Guangdong Academy of Agricultural Sciences, Guangzhou, 510640, China
| | - Zicong Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou, 510520, China
| | - Gengyuan Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou, 510520, China
| | - Linjun Hong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China.
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou, 510520, China.
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, South China Agricultural University, Guangzhou, 510642, China.
- Yunfu Subcenter of Guangdong Laboratory for Lingnan Modern Agriculture, Yunfu, 527300, China.
- Key Laboratory of South China Modern Biological Seed Industry, Ministry of Agriculture and Rural Affairs, Guangzhou, 510520, China.
| |
Collapse
|
45
|
Jácome-Ferrer P, Costas J. Exploring the causal effect of placental physiology in susceptibility to mental and addictive disorders: a Mendelian randomization study. Front Psychiatry 2024; 15:1396837. [PMID: 39135989 PMCID: PMC11317394 DOI: 10.3389/fpsyt.2024.1396837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/02/2024] [Indexed: 08/15/2024] Open
Abstract
Background Epidemiological studies have linked low birth weight to psychiatric disorders, including substance use disorders. Genomic analyses suggest a role of placental physiology on psychiatric risk. We investigated whether this association is causally related to impaired trophoblast function. Methods We conducted a two-sample summary-data Mendelian randomization study using as instrumental variables those genetic variants strongly associated with birth weight, whose effect is exerted through the fetal genome, and are located near genes with differential expression in trophoblasts. Eight psychiatric and substance use disorders with >10,000 samples were included as outcomes. The inverse variance weighted method was used as the main analysis and several sensitivity analyses were performed for those significant results. Results The inverse variance weighted estimate, based on 14 instrumental variables, revealed an association, after correction for multiple tests, between birth weight and broadly defined depression (β = -0.165, 95% CI = -0.282 to -0.047, P = 0.0059). Sensitivity analyses revealed the absence of heterogeneity in the effect of instrumental variables, confirmed by leave-one-out analysis, MR_Egger intercept, and MR_PRESSO. The effect was consistent using robust methods. Reverse causality was not detected. The effect was specifically linked to genetic variants near genes involved in trophoblast physiology instead of genes with fetal effect on birth weight or involved in placenta development. Conclusion Impaired trophoblast functioning, probably leading to reduced fetal brain oxygen and nutrient supply, is causally related to broadly defined depression. Considering the therapeutic potential of some agents to treat fetal growth restriction, further research on the effect of trophoblast physiology on mental disorders may have future implications in prevention.
Collapse
Affiliation(s)
- Pablo Jácome-Ferrer
- Psychiatric Genetics group, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Universidade de Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Javier Costas
- Psychiatric Genetics group, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Complexo Hospitalario Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saúde (SERGAS), Santiago de Compostela, Spain
| |
Collapse
|
46
|
Gunter-Rahman F, Mallett S, White F, Jacques PÉ, Raju RM, Hivert MF, Lee EA. Maternal obesity and offspring neurodevelopment are associated with hypoxic gene expression in term human placenta. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602900. [PMID: 39026777 PMCID: PMC11257614 DOI: 10.1101/2024.07.10.602900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
One third of women in the United States are affected by obesity during pregnancy. Maternal obesity (MO) is associated with an increased risk of neurodevelopmental and metabolic disorders in the offspring. The placenta, located at the maternal-fetal interface, is a key organ determining fetal development and likely contributes to programming of long-term offspring health. We profiled the term placental transcriptome in humans (pre-pregnancy BMI 35+ [MO condition] or 18.5-25 [lean condition]) using single-nucleus RNA-seq to compare expression profiles in MO versus lean conditions, and to reveal potential mechanisms underlying offspring disease risk. We recovered 62,864 nuclei of high quality from 10 samples each from the maternal-facing and fetal-facing sides of the placenta. On both sides in several cell types, MO was associated with upregulation of hypoxia response genes. On the maternal-facing side only, hypoxia gene expression was associated with offspring neurodevelopmental measures, in Gen3G, an independent pregnancy cohort with bulk placental tissue RNA-seq. We leveraged Gen3G to determine genes that correlated with impaired neurodevelopment and found these genes to be most highly expressed in extravillous trophoblasts (EVTs). EVTs further showed the strongest correlation between neurodevelopment impairment gene scores (NDIGSs) and the hypoxia gene score. We reanalyzed gene expression of cultured EVTs, and found increased NDIGSs associated with exposure to hypoxia. Among EVTs, accounting for the hypoxia gene score attenuated 44% of the association between BMI and NDIGSs. These data suggest that hypoxia in EVTs may be a key process in the neurodevelopmental programming of fetal exposure to MO.
Collapse
|
47
|
Umapathy A, Clark A, Sehgal A, Karanam V, Rajaraman G, Kalionis B, Jones H, James J, Murthi P. Molecular regulators of defective placental and cardiovascular development in fetal growth restriction. Clin Sci (Lond) 2024; 138:761-775. [PMID: 38904187 PMCID: PMC11193155 DOI: 10.1042/cs20220428] [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: 12/29/2023] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
Placental insufficiency is one of the major causes of fetal growth restriction (FGR), a significant pregnancy disorder in which the fetus fails to achieve its full growth potential in utero. As well as the acute consequences of being born too small, affected offspring are at increased risk of cardiovascular disease, diabetes and other chronic diseases in later life. The placenta and heart develop concurrently, therefore placental maldevelopment and function in FGR may have profound effect on the growth and differentiation of many organ systems, including the heart. Hence, understanding the key molecular players that are synergistically linked in the development of the placenta and heart is critical. This review highlights the key growth factors, angiogenic molecules and transcription factors that are common causes of defective placental and cardiovascular development.
Collapse
Affiliation(s)
- Anandita Umapathy
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Alys Clark
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
- Auckland Bioengineering Institute, Bioengineering Institute, New Zealand
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia and Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Vijaya Karanam
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
| | - Gayathri Rajaraman
- First year college, Victoria University, St Albans, Victoria 3021, Australia
| | - Bill Kalionis
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Victoria, Australia
| | - Helen N. Jones
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, U.S.A
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, U.S.A
| | - Jo James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
- Auckland Bioengineering Institute, Bioengineering Institute, New Zealand
| | - Padma Murthi
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Victoria, Australia
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
48
|
Bi S, Huang L, Chen Y, Hu Z, Li S, Wang Y, Huang B, Zhang L, Huang Y, Dai B, Du L, Tu Z, Wang Y, Xu D, Xu X, Sun W, Kzhyshkowska J, Wang H, Chen D, Wang F, Zhang S. KAT8-mediated H4K16ac is essential for sustaining trophoblast self-renewal and proliferation via regulating CDX2. Nat Commun 2024; 15:5602. [PMID: 38961108 PMCID: PMC11222414 DOI: 10.1038/s41467-024-49930-6] [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: 12/04/2023] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
Abnormal trophoblast self-renewal and differentiation during early gestation is the major cause of miscarriage, yet the underlying regulatory mechanisms remain elusive. Here, we show that trophoblast specific deletion of Kat8, a MYST family histone acetyltransferase, leads to extraembryonic ectoderm abnormalities and embryonic lethality. Employing RNA-seq and CUT&Tag analyses on trophoblast stem cells (TSCs), we further discover that KAT8 regulates the transcriptional activation of the trophoblast stemness marker, CDX2, via acetylating H4K16. Remarkably, CDX2 overexpression partially rescues the defects arising from Kat8 knockout. Moreover, increasing H4K16ac via using deacetylase SIRT1 inhibitor, EX527, restores CDX2 levels and promoted placental development. Clinical analysis shows reduced KAT8, CDX2 and H4K16ac expression are associated with recurrent pregnancy loss (RPL). Trophoblast organoids derived from these patients exhibit impaired TSC self-renewal and growth, which are significantly ameliorated with EX527 treatment. These findings suggest the therapeutic potential of targeting the KAT8-H4K16ac-CDX2 axis for mitigating RPL, shedding light on early gestational abnormalities.
Collapse
Affiliation(s)
- Shilei Bi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lijun Huang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yongjie Chen
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Zhenhua Hu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Shanze Li
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Yifan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Baoying Huang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lizi Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yuanyuan Huang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Beibei Dai
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Lili Du
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Zhaowei Tu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yijing Wang
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Dan Xu
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Xiaotong Xu
- National Institute of Biological Sciences, Beijing, 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China
| | - Wen Sun
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Julia Kzhyshkowska
- Institute of Transfusion Medicine and Immunology, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, 68167, Mannheim, Germany
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Dunjin Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Fengchao Wang
- National Institute of Biological Sciences, Beijing, 102206, China.
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, 102206, China.
| | - Shuang Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
- Guangdong-Hong Kong-Macao Great Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
49
|
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
|
50
|
Ozarslan N, Robinson JF, Buarpung S, Kim MY, Ansbro MR, Akram J, Montoya DJ, Kamya MR, Kakuru A, Dorsey G, Rosenthal PJ, Cheng G, Feeney ME, Fisher SJ, Gaw SL. Gravidity influences distinct transcriptional profiles of maternal and fetal placental macrophages at term. Front Immunol 2024; 15:1384361. [PMID: 38994356 PMCID: PMC11237841 DOI: 10.3389/fimmu.2024.1384361] [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: 02/09/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Introduction Maternal intervillous monocytes (MIMs) and fetal Hofbauer cells (HBCs) are myeloid-derived immune cells at the maternal-fetal interface. Maternal reproductive history is associated with differential risk of pregnancy complications. The molecular phenotypes and roles of these distinct monocyte/macrophage populations and the influence of gravidity on these phenotypes has not been systematically investigated. Methods Here, we used RNA sequencing to study the transcriptional profiles of MIMs and HBCs in normal term pregnancies. Results Our analyses revealed distinct transcriptomes of MIMs and HBCs. Genes involved in differentiation and cell organization pathways were more highly expressed in MIMs vs. HBCs. In contrast, HBCs had higher expression of genes involved in inflammatory responses and cell surface receptor signaling. Maternal gravidity influenced monocyte programming, as expression of pro-inflammatory molecules was significantly higher in MIMs from multigravidae compared to primigravidae. In HBCs, multigravidae displayed enrichment of gene pathways involved in cell-cell signaling and differentiation. Discussion Our results demonstrated that MIMs and HBCs have highly divergent transcriptional signatures, reflecting their distinct origins, locations, functions, and roles in inflammatory responses. Furthermore, maternal gravidity influences the gene signatures of MIMs and HBCs, potentially modulating the interplay between tolerance and trained immunity. The phenomenon of reproductive immune memory may play a novel role in the differential susceptibility of primigravidae to pregnancy complications.
Collapse
Affiliation(s)
- Nida Ozarslan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Joshua F. Robinson
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Sirirak Buarpung
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - M. Yvonne Kim
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Megan R. Ansbro
- Obstetrics & Gynecology Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Jason Akram
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Dennis J. Montoya
- Department of Molecular, Cellular & Developmental Biology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
- Department of Biochemistry and Molecular Medicine, University of California Davis Health, Sacramento, CA, United States
| | - Moses R. Kamya
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Department of Medicine, Makerere University, Kampala, Uganda
| | - Abel Kakuru
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Grant Dorsey
- Division of HIV, Global Medicine, and Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Philip J. Rosenthal
- Division of HIV, Global Medicine, and Infectious Diseases, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Genhong Cheng
- Department of Molecular Immunology and Genetics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Margaret E. Feeney
- Division of Experimental Medicine, Department of Medicine and Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Susan J. Fisher
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| | - Stephanie L. Gaw
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology & Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
- Center for Reproductive Sciences and Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California (UCSF), San Francisco, San Francisco, CA, United States
| |
Collapse
|