1
|
Cinkornpumin JK, Kwon SY, Prandstetter AM, Maxian T, Sirois J, Goldberg J, Zhang J, Saini D, Dasgupta P, Jeyarajah MJ, Renaud SJ, Paul S, Haider S, Pastor WA. Hypoxia and loss of GCM1 expression prevent differentiation and contact inhibition in human trophoblast stem cells. Stem Cell Reports 2025; 20:102481. [PMID: 40280139 DOI: 10.1016/j.stemcr.2025.102481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
During the first stages of embryonic development, the placenta develops under very low oxygen tension (∼1%-2% O2), so we sought to determine the regulatory role of oxygen in human trophoblast stem cells (hTSCs). We find that low oxygen promotes hTSC self-renewal but inhibits differentiation to syncytiotrophoblast (STB) and extravillous trophoblast (EVT). The transcription factor GCM1 (glial cell missing transcription factor 1) is downregulated in low oxygen, and concordantly, there is substantial reduction of GCM1-regulated genes in hypoxic conditions. Knockout of GCM1 in hTSC likewise impaired EVT and STB formation. Treatment with a phosphatidylinositol 3-kinase (PI3K) inhibitor reported to reduce GCM1 protein levels likewise counteracts spontaneous or directed differentiation. Additionally, chromatin immunoprecipitation of GCM1 showed binding near key genes upregulated upon differentiation including the contact inhibition factor CDKN1C. Loss of GCM1 resulted in downregulation of CDKN1C and corresponding loss of contact inhibition, implicating GCM1 in regulation of this critical process.
Collapse
Affiliation(s)
| | - Sin Young Kwon
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Anna-Maria Prandstetter
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Theresa Maxian
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - Jacinthe Sirois
- Department of Biochemistry, McGill University, Montreal, QC, Canada; The Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - James Goldberg
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Joy Zhang
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Deepak Saini
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Purbasa Dasgupta
- Department of Pathology and Laboratory Medicine, University of Kansas, Kansas City, Kansas, USA
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, University of Kansas, Kansas City, Kansas, USA; Institute for Reproduction and Developmental Sciences, University of Kansas, Kansas City, Kansas, USA; Department of Obstetrics and Gynecology, University of Kansas, Kansas City, Kansas, USA
| | - Sandra Haider
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Vienna, Austria
| | - William A Pastor
- Department of Biochemistry, McGill University, Montreal, QC, Canada; The Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
| |
Collapse
|
2
|
Meinhardt G, Waldhäusl H, Lackner AI, Wächter J, Maxian T, Höbler AL, Vondra S, Kunihs V, Saleh L, Haslinger P, Kiraly P, Szilagyi A, Than NG, Pollheimer J, Haider S, Knöfler M. The multifaceted roles of the transcriptional coactivator TAZ in extravillous trophoblast development of the human placenta. Proc Natl Acad Sci U S A 2025; 122:e2426385122. [PMID: 40228123 PMCID: PMC12037006 DOI: 10.1073/pnas.2426385122] [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: 12/17/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025] Open
Abstract
Insights into the molecular processes that drive early development of the human placenta is crucial for our understanding of pregnancy complications such as preeclampsia and fetal growth restriction, since defects in maturation of its epithelial cell, the trophoblast, have been detected in the severe forms of these diseases. However, key regulators specifying the differentiated trophoblast subtypes of the placenta are only slowly emerging. By using diverse trophoblast cell models, we herein show that the transcriptional coactivator of HIPPO signaling, TAZ, plays a pivotal role in the development of invasive extravillous trophoblasts (EVTs), cells that are essential for decidual vessel remodeling and adaption of maternal blood flow to the placenta. Ribonucleic acid sequencing (RNA-seq) or protein analyses upon TAZ gene silencing or CRISPR-Cas9-mediated knockout in differentiating trophoblast stem cells, organoids, primary EVTs, choriocarcinoma cells, or villous explant cultures unraveled that the coactivator promoted expression of genes associated with EVT identity, motility, and survival. Accordingly, depletion or chemical inhibition of TAZ, interacting with TEA domain family member 1 (TEAD1), impaired EVT differentiation, invasion, and migration and triggered apoptosis in the different trophoblast models. Notably, the coactivator also suppressed cell cycle genes and regulators of trophoblast self-renewal and prevented EVTs from cell fusion in organoids and primary cultures. Moreover, TAZ promoted human leukocyte antigen G (HLA-G) surface expression and increased NUAK1 kinase in EVTs thereby maintaining its own expression. In summary, the transcriptional coactivator TAZ plays a multifaceted role in the development of the EVT cell lineage by controlling different biological processes that initiate and preserve differentiation.
Collapse
Affiliation(s)
- Gudrun Meinhardt
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Hanna Waldhäusl
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Andreas I. Lackner
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Jasmin Wächter
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Theresa Maxian
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Anna-Lena Höbler
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Sigrid Vondra
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Victoria Kunihs
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Leila Saleh
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Peter Haslinger
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Peter Kiraly
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
| | - Andras Szilagyi
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
| | - Nandor G. Than
- Systems Biology of Reproduction Lendulet Group, Institute of Molecular Life Sciences, Hungarian Research Network (HUN-REN) Research Centre for Natural Sciences, Budapest1117, Hungary
- Maternity Private Clinic of Obstetrics and Gynecology, Budapest1126, Hungary
- Department of Obstetrics and Gynecology, Semmelweis University, Budapest1088, Hungary
| | - Jürgen Pollheimer
- Maternal-Fetal Immunology Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Sandra Haider
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| | - Martin Knöfler
- Placental Development Group, Reproductive Biology Unit, Department of Obstetrics and Gynaecology, Medical University of Vienna, ViennaA-1090, Austria
| |
Collapse
|
3
|
Keding LT, Heselton AR, Ren E, Shaw SA, Koenig MR, Golos TG, Schmidt JK. In vitro differentiation of macaque extravillous trophoblasts in a low oxygen environment. Placenta 2025; 163:16-28. [PMID: 40024139 PMCID: PMC11955297 DOI: 10.1016/j.placenta.2025.02.014] [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: 07/23/2024] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
INTRODUCTION Early primate placental development occurs within a low oxygen (O2) environment, whereas in vitro differentiation of trophoblasts is performed at supraphysiologic O2 levels. Macaque trophoblast stem cells (TSCs) are capable of differentiation to extravillous trophoblasts (EVTs) in vitro, yet the morphological heterogeneity observed across cells lines necessitates evaluation of optimal culture conditions. Our objectives were to determine the impact of oxygen on the in vitro differentiation of macaque TSCs and to refine the molecular characterization of TSC-differentiated EVTs. METHODS Macaque TSCs were differentiated to EVTs in either 20% or 5% O2. Gene and protein expression profiles were compared between TSCs and EVTs and between differentiation conditions. Immunohistochemical analysis was performed on early gestation macaque placental tissues to assess in vivo expression of Ki-67, NCAM1 and monkey chorionic gonadotropin (mCG). RESULTS EVTs differentiated in 20% O2 had significantly higher expression of CGA, CGB and NOTCH2 and decreased HIF1A expression compared to those cultured in 5% O2. Regardless of oxygen condition, nearly all EVTs expressed NCAM1 and Mamu-AG, the macaque-specific homolog of human EVT marker HLA-G. In vivo placental expression of NCAM1 was restricted to EVTs within the trophoblastic shell and endovasculature, revealing a macaque EVT marker within the placenta. DISCUSSION Reduced oxygen minimally impacted macaque EVT differentiation in vitro. Elevated expression of the endovascular EVT marker NOTCH2 potentially suggests that 20% O2 supported differentiation of more mature EVTs. Altogether, a standard 20% O2 environment supports macaque EVT differentiation in vitro and the results further validate the identity of macaque TSC-differentiated EVTs.
Collapse
Affiliation(s)
- Logan T Keding
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, 202 South Park St, Madison, WI, 53715, USA
| | - Avery R Heselton
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA
| | - Emily Ren
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA
| | - Sarah A Shaw
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA
| | - Michelle R Koenig
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA; Department of Obstetrics and Gynecology, University of Wisconsin-Madison, 202 South Park St, Madison, WI, 53715, USA; Department of Comparative Biosciences, School of Veterinary Medicine, 2015 Linden Dr, Madison, WI, 53706, USA
| | - Jenna K Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, 1223 Capitol Ct, Madison, WI, 53715, USA.
| |
Collapse
|
4
|
Shi X, Xi C, Dong B, Yan Z, Liu W, Gao S, Chen D. Maternal infection with SARS-CoV-2 during early pregnancy induces hypoxia at the maternal-fetal interface. Cell Prolif 2025; 58:e13749. [PMID: 39375953 PMCID: PMC11839197 DOI: 10.1111/cpr.13749] [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: 06/06/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic increases the risk of adverse fetal outcomes during pregnancy. Maternal infection during pregnancy, particularly with cytomegalovirus (CMV), hepatitis B and C virus, and human immunodeficiency virus can have detrimental effects on both mother and fetus, potentially leading to adverse outcomes such as spontaneous abortion or neonatal infection. However, the impact of severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection on the maternal-fetal interface remains poorly understood. In this study, we initially utilised immunofluorescence and immunohistochemical to investigate placental samples from pregnant women who were infected with SARS-CoV-2 during the first trimester. Our data indicate that infection in the first trimester induces an upregulation of hypoxia inducible factor (HIF) levels at the maternal-fetal interface. Subsequently, single-cell RNA sequencing and metabolomics sequencing analyses reveal alterations in maternal-fetal interface. Remarkably, immune cells exhibited low expression levels of HIF possibly associated with immune activation. Furthermore, our findings demonstrate a gradual reduction in transcriptome and metabolic changes as gestation progressed beyond 12-16 weeks compared to samples obtained at 6-8 weeks gestation. Overall, our study suggests that early-stage SARS-CoV-2 infection during the first trimester leads to severe hypoxia and aberrant cell metabolism at the maternal-fetal interface which gradually resolves as pregnancy progresses. Nevertheless, these abnormal changes may have long-term implications for maternal-fetal interface development.
Collapse
Affiliation(s)
- Xiaohui Shi
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Chenxiang Xi
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Baoxing Dong
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Zihui Yan
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Wenqiang Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Shaorong Gao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Di Chen
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangChina
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative MedicineZhejiang UniversityHangzhouChina
- State Key Laboratory of Biobased Transportation Fuel TechnologyZhejiang UniversityHainingZhejiangChina
| |
Collapse
|
5
|
Hu Y, Li S, Hong Y, Peng D. C3AR1 as a target for preeclampsia: from bioinformatics and network pharmacology to experimental validation. BMC Pregnancy Childbirth 2025; 25:94. [PMID: 39885406 PMCID: PMC11780793 DOI: 10.1186/s12884-025-07221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/23/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND Preeclampsia, characterized by hypertension and proteinuria during pregnancy, poses significant risks to both mother and fetus. The complement system's aberrant activation, notably the C3AR1, is important to the pathogenesis of preeclampsia, although the precise mechanisms are not fully understood. MATERIALS AND METHODS Utilizing the Comparative Toxicogenomics Database (CTD) and Molecular Signatures Database (MSigDB), we identified complement system targets associated with preeclampsia and environmental pollutants. Expression validation was conducted through the Gene Expression Omnibus (GEO) database. Molecular docking predicted interactions between BPA, PFOS, and C3AR1. Immunohistochemical staining of 80 placental tissues (40 early-onset preeclampsia and 40 healthy controls) confirmed C3AR1 expression and its clinical correlation. Integrated bioinformatics analyses revealed C3AR1's role in preeclampsia's molecular mechanisms. Functional verification was assessed by knocking down C3AR1 in HTR-8/Svneo cells, including cell proliferation, invasion, and apoptosis. RESULTS Network pharmacology established connections between pollutants and preeclampsia, with C3AR1 as a key target. Molecular docking confirmed BPA and PFOS binding to C3AR1. Reduced C3AR1 in preeclamptic placentas correlated with maternal blood pressure, and showed high diagnostic potential (AUC = 0.95). C3AR1's involvement in preeclampsia was linked to Jak-STAT, TGF-β, and HIF-1 pathways, and associated with NK cell and M1 macrophage activity. C3AR1 knockdown in HTR-8/Svneo cells decreased proliferation and invasion, and increased apoptosis. CONCLUSION C3AR1 expression is diminished in preeclampsia placental tissues, correlating with disease severity, suggesting its potential as a biomarker. It is crucial for cellular functions and inflammation, with future studies aiming to leverage this for novel preeclampsia treatments. CLINICAL TRIAL Not applicable.
Collapse
Affiliation(s)
- Yongfeng Hu
- Obstetrics and Gynecology Center, Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
- Department of Obstetrics, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528308, China
| | - Shaoqiang Li
- Department of Obstetrics, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528308, China
| | - Yunhui Hong
- Department of Obstetrics, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, Guangdong, 528308, China
| | - Dongxian Peng
- Obstetrics and Gynecology Center, Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China.
| |
Collapse
|
6
|
Di Giorgio E, Xodo S, Orsaria M, Mariuzzi L, Picco R, Tolotto V, Cortolezzis Y, D'Este F, Grandi N, Driul L, Londero A, Xodo LE. The central role of creatine and polyamines in fetal growth restriction. FASEB J 2024; 38:e70222. [PMID: 39614665 DOI: 10.1096/fj.202401946r] [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: 08/20/2024] [Revised: 10/23/2024] [Accepted: 11/19/2024] [Indexed: 12/01/2024]
Abstract
Placental insufficiency often correlates with fetal growth restriction (FGR), a condition that has both short- and long-term effects on the health of the newborn. In our study, we analyzed placental tissue from infants with FGR and from infants classified as small for gestational age (SGA) or appropriate for gestational age (AGA), performing comprehensive analyses that included transcriptomics and metabolomics. By examining villus tissue biopsies and 3D trophoblast organoids, we identified significant metabolic changes in placentas associated with FGR. These changes include adaptations to reduced oxygen levels and modifications in arginine metabolism, particularly within the polyamine and creatine phosphate synthesis pathways. Specifically, we found that placentas with FGR utilize arginine to produce phosphocreatine, a crucial energy reservoir for ATP production that is essential for maintaining trophoblast function. In addition, we found polyamine insufficiency in FGR placentas due to increased SAT1 expression. SAT1 facilitates the acetylation and subsequent elimination of spermine and spermidine from trophoblasts, resulting in a deficit of polyamines that cannot be compensated by arginine or polyamine supplementation alone, unless SAT1 expression is suppressed. Our study contributes significantly to the understanding of metabolic adaptations associated with placental dysfunction and provides valuable insights into potential therapeutic opportunities for the future.
Collapse
Affiliation(s)
| | - Serena Xodo
- Clinic of Obstetrics and Gynecology, Santa Maria della Misericordia Hospital, ASUFC, Udine, Italy
| | - Maria Orsaria
- Institute of Pathology, Department of Medicine, University of Udine, Udine, Italy
| | - Laura Mariuzzi
- Institute of Pathology, Department of Medicine, University of Udine, Udine, Italy
| | | | | | | | | | - Nicole Grandi
- Laboratory of Molecular Virology, Department of Life and Environmental Sciences, University of Cagliari, Cagliari, Italy
| | - Lorenza Driul
- Department of Medicine, University of Udine, Udine, Italy
- Clinic of Obstetrics and Gynecology, Santa Maria della Misericordia Hospital, ASUFC, Udine, Italy
| | - Ambrogio Londero
- Obstetrics and Gynecology Unit, IRCCS Institute Giannina Gaslini, Genova, Italy
| | - Luigi E Xodo
- Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
7
|
Procházková K, Uhlík J. Influence of Hypoxia on the Airway Epithelium. Physiol Res 2024; 73:S557. [PMID: 39589303 PMCID: PMC11627265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 06/26/2024] [Indexed: 11/27/2024] Open
Abstract
The necessity of oxygen for metabolic processes means that hypoxia can lead to serious cell and tissue damage. On the other hand, in some situations, hypoxia occurs under physiological conditions and serves as an important regulation factor. The airway epithelium is specific in that it gains oxygen not only from the blood supply but also directly from the luminal air. Many respiratory diseases are associated with airway obstruction or excessive mucus production thus leading to luminal hypoxia. The main goal of this review is to point out how the airway epithelium reacts to hypoxic conditions. Cells detect low oxygen levels using molecular mechanisms involving hypoxia-inducible factors (HIFs). In addition, the cells of the airway epithelium appear to overexpress HIFs in hypoxic conditions. HIFs then regulate many aspects of epithelial cell functions. The effects of hypoxia include secretory cell stimulation and hyperplasia, epithelial barrier changes, and ciliogenesis impairment. All the changes can impair mucociliary clearance, exacerbate infection, and promote inflammation leading to damage of airway epithelium and subsequent airway wall remodeling. The modulation of hypoxia regulatory mechanisms may be one of the strategies for the treatment of obstructive respiratory diseases or diseases with mucus hyperproduction. Keywords: Secretory cells, Motile cilia, Epithelial barrier, Oxygenation, Obstructive respiratory diseases.
Collapse
Affiliation(s)
- K Procházková
- Department of Histology and Embryology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | |
Collapse
|
8
|
Lai Y, Fu Z, Gao Y, Ma N, Li L. Hypoxia-inducible factors (HIFs) in early pregnancy: implications for miscarriage†. Biol Reprod 2024; 111:987-999. [PMID: 39325972 DOI: 10.1093/biolre/ioae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 09/07/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024] Open
Abstract
Miscarriage poses a significant threat to both maternal and fetal health. Its etiology remains unknown, and there are no established effective identification or prevention strategies. A low-oxygen environment in early pregnancy is a physiological necessity for embryonic and placental growth. Hypoxia-inducible factors are a family of classic hypoxia signaling molecules whose expression level may fluctuate abnormally because of an imbalance in oxygen levels. Its unusual fluctuations initiate multiple signaling pathways at the maternal womb. Hypoxia-inducible factors are a family of classic hypoxia-signaling molecules and immune tolerance. Notably, aberrant regulation of these processes may lead to miscarriage. This review aims to clarify how the hypoxia-inducible factor-1α mediates the aberrant regulation of biological processes, including autophagy, metabolic reprogramming, et al., and how these effects impact trophoblasts and other cells at the maternal-fetal interface. These findings provide new insights into potential therapeutic and preventive strategies for miscarriage.
Collapse
Affiliation(s)
- Yuxuan Lai
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Zhiyu Fu
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Yaxin Gao
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Ning Ma
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| | - Lu Li
- Department of Social Medicine and Health Care Management, School of Public Health, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Environment and Population Health, School of Public Health, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
9
|
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
|
10
|
Cinkornpumin JK, Kwon SY, Prandstetter AM, Maxian T, Sirois J, Goldberg J, Zhang J, Saini D, Dasgupta P, Jeyarajah MJ, Renaud SJ, Paul S, Haider S, Pastor WA. Hypoxia and loss of GCM1 expression prevents differentiation and contact inhibition in human trophoblast stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612343. [PMID: 39314437 PMCID: PMC11419009 DOI: 10.1101/2024.09.10.612343] [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: 09/25/2024]
Abstract
The placenta develops alongside the embryo and nurtures fetal development to term. During the first stages of embryonic development, due to low blood circulation, the blood and ambient oxygen supply is very low (~1-2% O2) and gradually increases upon placental invasion. While a hypoxic environment is associated with stem cell self-renewal and proliferation, persistent hypoxia may have severe effects on differentiating cells and could be the underlying cause of placental disorders. We find that human trophoblast stem cells (hTSC) thrive in low oxygen, whereas differentiation of hTSC to trophoblast to syncytiotrophoblast (STB) and extravillous trophoblast (EVT) is negatively affected by hypoxic conditions. The pro-differentiation factor GCM1 (human Glial Cell Missing-1) is downregulated in low oxygen, and concordantly there is substantial reduction of GCM1-regulated genes in hypoxic conditions. Knockout of GCM1 in hTSC caused impaired EVT and STB formation and function, reduced expression of differentiation-responsive genes, and resulted in maintenance of self-renewal genes. Treatment with a PI3K inhibitor reported to reduce GCM1 protein levels likewise counteracts spontaneous or directed differentiation. Additionally, chromatin immunoprecipitation of GCM1 showed enrichment of GCM1-specific binding near key transcription factors upregulated upon differentiation including the contact inhibition factor CDKN1C. Loss of GCM1 resulted in downregulation of CDKN1C and corresponding loss of contact inhibition, implicating GCM1 in regulation of this critical process.
Collapse
Affiliation(s)
| | - Sin Young Kwon
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Anna-Maria Prandstetter
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Austria
| | - Theresa Maxian
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Austria
| | - Jacinthe Sirois
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- The Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - James Goldberg
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Joy Zhang
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Deepak Saini
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Purbasa Dasgupta
- Department of Pathology and Laboratory Medicine, University of Kansas, Kansas City, United States
| | - Mariyan J Jeyarajah
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario
| | - Stephen J Renaud
- Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario
| | - Soumen Paul
- Department of Pathology and Laboratory Medicine, University of Kansas, Kansas City, United States
- Institute for Reproduction and Developmental Sciences, University of Kansas, Kansas City, United States
- Department of Obstetrics and Gynecology, University of Kansas, Kansas City, United States
| | - Sandra Haider
- Placental Development Group, Reproductive Biology Unit, Medical University of Vienna, Austria
| | - William A Pastor
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
- The Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
11
|
Doria-Borrell P, Pérez-García V. Understanding the intersection between placental development and cancer: Lessons from the tumor suppressor BAP1. Commun Biol 2024; 7:1053. [PMID: 39191942 PMCID: PMC11349880 DOI: 10.1038/s42003-024-06689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
The placenta, a pivotal organ in mammalian reproduction, allows nutrient exchange and hormonal signaling between the mother and the developing fetus. Understanding its molecular intricacies is essential for deciphering normal embryonic development and pathological conditions such as tumorigenesis. Here, we explore the multifaceted role of the tumor suppressor BRCA1-associated protein 1 (BAP1) in cancer and placentation. Initially recognized for its tumor-suppressive properties, BAP1 has emerged as a key regulator at the intersection of tumorigenesis and placental development. BAP1 influences crucial cellular processes such as cell death, proliferation, metabolism, and response to hypoxic conditions. By integrating insights from tumor and developmental biology, we illuminate the complex molecular pathways orchestrated by BAP1. This perspective highlights BAP1's significant impact on both cancer and placental development, and suggests novel therapeutic strategies that could improve outcomes for pregnancy disorders and cancer.
Collapse
Affiliation(s)
| | - Vicente Pérez-García
- Centro de Investigación Príncipe Felipe, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| |
Collapse
|
12
|
Swingle KL, Hamilton AG, Mitchell MJ. Flow cytometric analysis of the murine placenta to evaluate nanoparticle platforms during pregnancy. Placenta 2024:S0143-4004(24)00618-0. [PMID: 39181830 PMCID: PMC11822046 DOI: 10.1016/j.placenta.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Clinically approved therapeutics for obstetric conditions are extremely limited, with over 80% of drugs lacking appropriate labeling information for pregnant individuals. The pathology for many of these obstetric conditions can be linked to the placenta, necessitating the development of therapeutic platforms for selective drug delivery to the placenta. When evaluating therapeutics for placental delivery, literature has focused on ex vivo delivery to human placental cells and tissue, which can be difficult to source for non-clinical researchers. Evaluating in vivo drug delivery to the placenta using small animal models can be more accessible than using human tissue, but robust, quantitative methods to characterize delivery remain poorly established. Here, we report a flow cytometric method to evaluate in vivo drug delivery to the murine placenta. Specifically, we describe techniques to identify key cell types in the murine placenta - trophoblasts, endothelial cells, and immune cells - via flow cytometric analysis. While we have employed this method to detect lipid nanoparticle-mediated nucleic acid delivery, this approach can extend to a variety of drug carriers (e.g., liposomes, exosomes, polymeric and metallic nanoparticles) and payloads (e.g., small molecules, proteins, other nucleic acids). Similarly, we describe the application of this method toward immunophenotypic analysis to assess changes in the placental immune environment during disease or in response to a therapeutic. Together, the techniques reported herein aim to broaden the accessibility of placental research in an effort to encourage collaboration between physician-scientists, engineers, placental biologists, and clinicians for developing novel therapeutics to treat placental conditions during pregnancy.
Collapse
Affiliation(s)
- Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Alex G Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for RNA Innovation, Perelman School of Medicine, Philadelphia, PA, United States; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
13
|
Morey R, Soncin F, Kallol S, Sah N, Manalo Z, Bui T, Slamecka J, Cheung VC, Pizzo D, Requena DF, Chang CW, Farah O, Kittle R, Meads M, Horii M, Fisch K, Parast MM. Single-cell transcriptomics reveal differences between chorionic and basal plate cytotrophoblasts and trophoblast stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.12.603155. [PMID: 39071344 PMCID: PMC11275976 DOI: 10.1101/2024.07.12.603155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Cytotrophoblast (CTB) of the early gestation human placenta are bipotent progenitor epithelial cells, which can differentiate into invasive extravillous trophoblast (EVT) and multinucleated syncytiotrophoblast (STB). Trophoblast stem cells (TSC), derived from early first trimester placentae, have also been shown to be bipotential. In this study, we set out to probe the transcriptional diversity of first trimester CTB and compare TSC to various subgroups of CTB. We performed single-cell RNA sequencing on six normal placentae, four from early (6-8 weeks) and two from late (12-14 weeks) first trimester, of which two of the early first trimester cases were separated into basal (maternal) and chorionic (fetal) fractions prior to sequencing. We also sequenced three TSC lines, derived from 6-8 week placentae, to evaluate similarities and differences between primary CTB and TSC. CTB clusters displayed notable distinctions based on gestational age, with early first trimester placentae showing enrichment for specific CTB subtypes, further influenced by origin from the basal or chorionic plate. Differential expression analysis of CTB from basal versus chorionic plate highlighted pathways associated with proliferation, unfolded protein response, and oxidative phosphorylation. We identified trophoblast states representing initial progenitor CTB, precursor STB, precursor and mature EVT, and multiple CTB subtypes. CTB progenitors were enriched in early first trimester placentae, with basal plate cells biased toward EVT, and chorionic plate cells toward STB, precursors. Clustering and trajectory inference analysis indicated that TSC were most like EVT precursor cells, with only a small percentage of TSC on the pre-STB differentiation trajectory. This was confirmed by flow cytometric analysis of 6 different TSC lines, which showed uniform expression of proximal column markers ITGA2 and ITGA5. Additionally, we found that ITGA5+ CTB could be plated in 2D, forming only EVT upon spontaneous differentiation, but failed to form self-renewing organoids; conversely, ITGA5-CTB could not be plated in 2D, but readily formed organoids. Our findings suggest that distinct CTB states exist in different regions of the placenta as early as six weeks gestation and that current TSC lines most closely resemble ITGA5+ CTB, biased toward the EVT lineage.
Collapse
Affiliation(s)
- Robert Morey
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Francesca Soncin
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Sampada Kallol
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Nirvay Sah
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Zoe Manalo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tony Bui
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jaroslav Slamecka
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Virginia Chu Cheung
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Don Pizzo
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Daniela F Requena
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ching-Wen Chang
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
| | - Omar Farah
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
| | - Ryan Kittle
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Morgan Meads
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mariko Horii
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kathleen Fisch
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Mana M Parast
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
- Center for Perinatal Discovery, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
14
|
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
|
15
|
Donohoe ME, Morey R, Li Y, Pizzo D, Kallol S, Cho HY, Soncin F, Parast MM. Identification of HTRA4 as a Transcriptional Target of p63 in Trophoblast. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1162-1170. [PMID: 38880601 PMCID: PMC11220921 DOI: 10.1016/j.ajpath.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 06/18/2024]
Abstract
The placenta plays a crucial role in pregnancy success. ΔNp63α (p63), a transcription factor from the TP53 family, is highly expressed in villous cytotrophoblasts (CTBs), the epithelial stem cells of the human placenta, and is involved in CTB maintenance and differentiation. We examined the mechanisms of action of p63 by identifying its downstream targets. Gene expression changes were evaluated following overexpression and knockdown of p63 in the JEG3 choriocarcinoma cell line, using microarray-based RNA profiling. High-temperature requirement A4 (HTRA4), a placenta-specific serine protease involved in trophoblast differentiation and altered in preeclampsia, was identified as a gene reciprocally regulated by p63, and its expression was characterized in primary human placental tissues by RNA-sequencing and in situ hybridization. Potential p63 DNA-binding motifs were identified in the HTRA4 promoter, and p63 occupancy at some of these sites was confirmed using chromatin immunoprecipitation, followed by quantitative PCR in both JEG3 and trophoblast stem cells. These data begin to identify members of the transcriptional network downstream of p63, thus laying the groundwork for probing mechanisms by which this important transcription factor regulates trophoblast stemness and differentiation.
Collapse
Affiliation(s)
- Mary E Donohoe
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Robert Morey
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Yingchun Li
- Center for Genes, Environment, and Health, National Jewish Health, Denver, Colorado
| | - Donald Pizzo
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Sampada Kallol
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Hee-Young Cho
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
16
|
Yan X, Fang Y, Yuan Y, Ding Y, Yu H, Li Y, Shi Q, Gao Y, Zhou X, Zhang D, Yuan E, Zhou H, Zhao X, Zhang L. Combined analysis of the effects of hypoxia and oxidative stress on DNA methylation and the transcriptome in HTR-8/SVneo trophoblast cells. J Cell Mol Med 2024; 28:e18469. [PMID: 38899809 PMCID: PMC11187809 DOI: 10.1111/jcmm.18469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
The alterations in DNA methylation and transcriptome in trophoblast cells under conditions of low oxygen and oxidative stress have major implications for pregnancy-related disorders. However, the exact mechanism is still not fully understood. In this study, we established models of hypoxia (H group) and oxidative stress (HR group) using HTR-8/SVneo trophoblast cells and performed combined analysis of genome-wide DNA methylation changes using reduced representation bisulphite sequencing and transcriptome expression changes using RNA sequencing. Our findings revealed that the H group exhibited a higher number of differentially methylated genes and differentially expressed genes than the HR group. In the H group, only 0.90% of all differentially expressed genes displayed simultaneous changes in DNA methylation and transcriptome expression. After the threshold was expanded, this number increased to 6.29% in the HR group. Notably, both the H group and HR group exhibited concurrent alterations in DNA methylation and transcriptome expression within Axon guidance and MAPK signalling pathway. Among the top 25 differentially methylated KEGG pathways in the promoter region, 11 pathways were commonly enriched in H group and HR group, accounting for 44.00%. Among the top 25 KEGG pathways in transcriptome with significant differences between the H group and HR group, 10 pathways were consistent, accounting for 40.00%. By integrating our previous data on DNA methylation from preeclamptic placental tissues, we identified that the ANKRD37 and PFKFB3 genes may contribute to the pathogenesis of preeclampsia through DNA methylation-mediated transcriptome expression under hypoxic conditions.
Collapse
Affiliation(s)
- Xinjing Yan
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
| | - Yang Fang
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
| | - Yujie Yuan
- Judicial Appraisal InstitutionThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Yangnan Ding
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
| | - Haiyang Yu
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
| | - Yina Li
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
| | - Qianqian Shi
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
| | - Yongrui Gao
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
| | - Xinyuan Zhou
- Department of Obstetrics and GynecologyThe Third Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Dongxin Zhang
- Microbiome Medicine Center, Department of Laboratory MedicineZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Enwu Yuan
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
- Tianjian Advanced Biomedical LaboratoryZhengzhouChina
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory MedicineZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Xin Zhao
- Tianjian Advanced Biomedical LaboratoryZhengzhouChina
| | - Linlin Zhang
- Department of Laboratory MedicineThird Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
- Zhengzhou Key Laboratory for In Vitro Diagnosis of Hypertensive Disorders of PregnancyZhengzhouChina
- Tianjian Advanced Biomedical LaboratoryZhengzhouChina
| |
Collapse
|
17
|
Shannon MJ, McNeill GL, Koksal B, Baltayeva J, Wächter J, Castellana B, Peñaherrera MS, Robinson WP, Leung PCK, Beristain AG. Single-cell assessment of primary and stem cell-derived human trophoblast organoids as placenta-modeling platforms. Dev Cell 2024; 59:776-792.e11. [PMID: 38359834 DOI: 10.1016/j.devcel.2024.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/15/2023] [Accepted: 01/23/2024] [Indexed: 02/17/2024]
Abstract
Human trophoblast stem cells (hTSCs) and related trophoblast organoids are state-of-the-art culture systems that facilitate the study of trophoblast development and human placentation. Using single-cell transcriptomics, we evaluate how organoids derived from freshly isolated first-trimester trophoblasts or from established hTSC cell lines reproduce developmental cell trajectories and transcriptional regulatory processes defined in vivo. Although organoids from primary trophoblasts and hTSCs overall model trophoblast differentiation with accuracy, specific features related to trophoblast composition, trophoblast differentiation, and transcriptional drivers of trophoblast development show levels of misalignment. This is best illustrated by the identification of an expanded progenitor state in stem cell-derived organoids that is nearly absent in vivo and transcriptionally shares both villous cytotrophoblast and extravillous trophoblast characteristics. Together, this work provides a comprehensive resource that identifies strengths and limitations of current trophoblast organoid platforms.
Collapse
Affiliation(s)
- Matthew J Shannon
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Gina L McNeill
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Burak Koksal
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Jennet Baltayeva
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Jasmin Wächter
- Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Barbara Castellana
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Maria S Peñaherrera
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Wendy P Robinson
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Peter C K Leung
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada
| | - Alexander G Beristain
- The British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Obstetrics & Gynecology, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
18
|
Kim M, Jang YJ, Lee M, Guo Q, Son AJ, Kakkad NA, Roland AB, Lee BK, Kim J. The transcriptional regulatory network modulating human trophoblast stem cells to extravillous trophoblast differentiation. Nat Commun 2024; 15:1285. [PMID: 38346993 PMCID: PMC10861538 DOI: 10.1038/s41467-024-45669-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 01/31/2024] [Indexed: 02/15/2024] Open
Abstract
During human pregnancy, extravillous trophoblasts play crucial roles in placental invasion into the maternal decidua and spiral artery remodeling. However, regulatory factors and their action mechanisms modulating human extravillous trophoblast specification have been unknown. By analyzing dynamic changes in transcriptome and enhancer profile during human trophoblast stem cell to extravillous trophoblast differentiation, we define stage-specific regulators, including an early-stage transcription factor, TFAP2C, and multiple late-stage transcription factors. Loss-of-function studies confirm the requirement of all transcription factors identified for adequate differentiation, and we reveal that the dynamic changes in the levels of TFAP2C are essential. Notably, TFAP2C pre-occupies the regulatory elements of the inactive extravillous trophoblast-active genes during the early stage of differentiation, and the late-stage transcription factors directly activate extravillous trophoblast-active genes, including themselves as differentiation further progresses, suggesting sequential actions of transcription factors assuring differentiation. Our results reveal stage-specific transcription factors and their inter-connected regulatory mechanisms modulating extravillous trophoblast differentiation, providing a framework for understanding early human placentation and placenta-related complications.
Collapse
Affiliation(s)
- Mijeong Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Yu Jin Jang
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Muyoung Lee
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Qingqing Guo
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Albert J Son
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Nikita A Kakkad
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Abigail B Roland
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Bum-Kyu Lee
- Department of Biomedical Sciences, Cancer Research Center, University at Albany, State University of New York, Rensselaer, NY, 12144, USA
| | - Jonghwan Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA.
| |
Collapse
|
19
|
Xie YP, Lin S, Xie BY, Zhao HF. Recent progress in metabolic reprogramming in gestational diabetes mellitus: a review. Front Endocrinol (Lausanne) 2024; 14:1284160. [PMID: 38234430 PMCID: PMC10791831 DOI: 10.3389/fendo.2023.1284160] [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: 08/28/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024] Open
Abstract
Gestational diabetes mellitus is a prevalent metabolic disease that can impact the normal course of pregnancy and delivery, leading to adverse outcomes for both mother and child. Its pathogenesis is complex and involves various factors, such as insulin resistance and β-cell dysfunction. Metabolic reprogramming, which involves mitochondrial oxidative phosphorylation and glycolysis, is crucial for maintaining human metabolic balance and is involved in the pathogenesis and progression of gestational diabetes mellitus. However, research on the link and metabolic pathways between metabolic reprogramming and gestational diabetes mellitus is limited. Therefore, we reviewed the relationship between metabolic reprogramming and gestational diabetes mellitus to provide new therapeutic strategies for maternal health during pregnancy and reduce the risk of developing gestational diabetes mellitus.
Collapse
Affiliation(s)
- Ya-ping Xie
- Nursing Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Bao-yuan Xie
- Nursing Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Hui-fen Zhao
- Nursing Department, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
20
|
Abstract
During placentation, villous cytotrophoblast (CTB) stem cells proliferate and fuse, giving rise to the multinucleated syncytiotrophoblast (STB), which represents the terminally differentiated villous layer as well as the maternal-fetal interface. The syncytiotrophoblast is at the forefront of nutrient, gas, and waste exchange while also harboring essential endocrine functions to support pregnancy and fetal development. Considering that mitochondrial dynamics and respiration have been implicated in stem cell fate decisions of several cell types and that the placenta is a mitochondria-rich organ, we will highlight the role of mitochondria in facilitating trophoblast differentiation and maintaining trophoblast function. We discuss both the process of syncytialization and the distinct metabolic characteristics associated with CTB and STB sub-lineages prior to and during syncytialization. As mitochondrial respiration is tightly coupled to redox homeostasis, we emphasize the adaptations of mitochondrial respiration to the hypoxic placental environment. Furthermore, we highlight the critical role of mitochondria in conferring the steroidogenic potential of the STB following differentiation. Ultimately, mitochondrial function and morphological changes centrally regulate respiration and influence trophoblast fate decisions through the production of reactive oxygen species (ROS), whose levels modulate the transcriptional activation or suppression of pluripotency or commitment genes.
Collapse
Affiliation(s)
- Tina Podinić
- Department of Pediatrics and Graduate Program in Medical Sciences, McMaster University, Hamilton, ON, Canada
| | - Andie MacAndrew
- Department of Pediatrics and Graduate Program in Medical Sciences, McMaster University, Hamilton, ON, Canada
| | - Sandeep Raha
- Department of Pediatrics and Graduate Program in Medical Sciences, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
21
|
Brumbaugh J, Aguado BA, Lysaght T, Goldstein LSB. Human fetal tissue is critical for biomedical research. Stem Cell Reports 2023; 18:2300-2312. [PMID: 37977142 PMCID: PMC10724055 DOI: 10.1016/j.stemcr.2023.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023] Open
Abstract
Human fetal tissue and cells derived from fetal tissue are crucial for biomedical research. Fetal tissues and cells are used to study both normal development and developmental disorders. They are broadly applied in vaccine development and production. Further, research using cells from fetal tissue is instrumental for studying many infectious diseases, including a broad range of viruses. These widespread applications underscore the value of fetal tissue research and reflect an important point: cells derived from fetal tissues have capabilities that cells from other sources do not. In many cases, increased functionality of cells derived from fetal tissues arises from increased proliferative capacity, ability to survive in culture, and developmental potential that is attenuated in adult tissues. This review highlights important, representative applications of fetal tissue for science and medicine.
Collapse
Affiliation(s)
- Justin Brumbaugh
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, CO, USA; University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, CO, USA; Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Brian A Aguado
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Tamra Lysaght
- Centre for Biomedical Ethics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lawrence S B Goldstein
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA; Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
22
|
Dietrich B, Kunihs V, Lackner AI, Meinhardt G, Koo BK, Pollheimer J, Haider S, Knöfler M. NOTCH3 signalling controls human trophoblast stem cell expansion and differentiation. Development 2023; 150:dev202152. [PMID: 37905445 DOI: 10.1242/dev.202152] [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: 07/03/2023] [Accepted: 10/19/2023] [Indexed: 11/02/2023]
Abstract
Failures in growth and differentiation of the early human placenta are associated with severe pregnancy disorders such as pre-eclampsia and fetal growth restriction. However, regulatory mechanisms controlling development of placental epithelial cells, the trophoblasts, remain poorly elucidated. Using trophoblast stem cells (TSCs), trophoblast organoids (TB-ORGs) and primary cytotrophoblasts (CTBs) of early pregnancy, we herein show that autocrine NOTCH3 signalling controls human placental expansion and differentiation. The NOTCH3 receptor was specifically expressed in proliferative CTB progenitors and its active form, the nuclear NOTCH3 intracellular domain (NOTCH3-ICD), interacted with the transcriptional co-activator mastermind-like 1 (MAML1). Doxycycline-inducible expression of dominant-negative MAML1 in TSC lines provoked cell fusion and upregulation of genes specific for multinucleated syncytiotrophoblasts, which are the differentiated hormone-producing cells of the placenta. However, progenitor expansion and markers of trophoblast stemness and proliferation were suppressed. Accordingly, inhibition of NOTCH3 signalling diminished growth of TB-ORGs, whereas overexpression of NOTCH3-ICD in primary CTBs and TSCs showed opposite effects. In conclusion, the data suggest that canonical NOTCH3 signalling plays a key role in human placental development by promoting self-renewal of CTB progenitors.
Collapse
Affiliation(s)
- Bianca Dietrich
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Victoria Kunihs
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Andreas I Lackner
- Maternal-Fetal Immunology Group, Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, A-1090 Vienna, Austria
| | - Gudrun Meinhardt
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Yuseong-Gu, Daejeon 34126, Republic of Korea
| | - Jürgen Pollheimer
- Maternal-Fetal Immunology Group, Department of Obstetrics and Gynecology, Reproductive Biology Unit, Medical University of Vienna, A-1090 Vienna, Austria
| | - Sandra Haider
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| | - Martin Knöfler
- Placental Development Group, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
23
|
Yokouchi-Konishi T, Liu Y, Feng L. Progesterone receptor membrane component 2 is critical for human placental extravillous trophoblast invasion. Biol Reprod 2023; 109:759-771. [PMID: 37665239 DOI: 10.1093/biolre/ioad109] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/14/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023] Open
Abstract
Proper extravillous trophoblast invasion is essential for normal placentation and pregnancy. However, the molecular mechanisms by which cytotrophoblasts differentiate into extravillous trophoblast are unclear. We discovered that in the first-trimester placenta, progesterone receptor membrane component 2 was highly expressed in syncytiotrophoblast but significantly lower in extravillous trophoblast and cytotrophoblasts, indicating a divergent role for progesterone receptor membrane component 2 in trophoblast functions. We aim to examine the role of progesterone receptor membrane component 2 in extravillous trophoblasts invasion mediated by both intracellular and extracellular signals. Progesterone receptor membrane component 2 knockdown and overexpression cells were established in HTR8/SVneo cells, a first-trimester extravillous trophoblast-derived cell model, by transfection with small-interfering RNA or progesterone receptor membrane component 2 plasmids, respectively. Progesterone receptor membrane component 2 knockdown led to cellular morphological changes , enhanced trophoblast proliferation,invasion, and promoted tube formation. These effects were mediated by the activation of hypoxia-inducible factor 1alpha and an increased expression of vascular endothelial growth factor A. The culture supernatant collected from progesterone receptor membrane component 2 knockdown cells did not significantly affect extravillous trophoblast invasion compared to the controls, indicating that extracellular signaling did not robustly regulate extravillous trophoblast invasion in this study. In conclusion, attenuation of progesterone receptor membrane component 2 plays a role in placentation by promoting cell proliferation, invasion, and angiogenesis in extravillous trophoblasts via activation of hypoxia-inducible factor 1 alpha signaling. We thus identified a new function of progesterone receptor membrane component 2 and provide insights on understanding the mechanisms of trophoblast invasion.
Collapse
Affiliation(s)
- Tae Yokouchi-Konishi
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
- Department of Obstetrics and Gynecology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Yongjie Liu
- Ministry of Education and Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
24
|
Wątroba M, Szewczyk G, Szukiewicz D. The Role of Sirtuin-1 (SIRT1) in the Physiology and Pathophysiology of the Human Placenta. Int J Mol Sci 2023; 24:16210. [PMID: 38003402 PMCID: PMC10671790 DOI: 10.3390/ijms242216210] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Sirtuins, especially SIRT1, play a significant role in regulating inflammatory response, autophagy, and cell response to oxidative stress. Since their discovery, sirtuins have been regarded as anti-ageing and longevity-promoting enzymes. Sirtuin-regulated processes seem to participate in the most prevalent placental pathologies, such as pre-eclampsia. Furthermore, more and more research studies indicate that SIRT1 may prevent pre-eclampsia development or at least alleviate its manifestations. Having considered this, we reviewed recent studies on the role of sirtuins, especially SIRT1, in processes determining normal or abnormal development and functioning of the placenta.
Collapse
Affiliation(s)
| | | | - Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Medical University of Warsaw, Chałubinskiego 5, 02-004 Warsaw, Poland; (M.W.); (G.S.)
| |
Collapse
|
25
|
Sugimoto J, Schust DJ, Sugimoto M, Jinno Y, Kudo Y. Controlling Trophoblast Cell Fusion in the Human Placenta-Transcriptional Regulation of Suppressyn, an Endogenous Inhibitor of Syncytin-1. Biomolecules 2023; 13:1627. [PMID: 38002309 PMCID: PMC10668956 DOI: 10.3390/biom13111627] [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: 08/14/2023] [Revised: 10/24/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Cell fusion in the placenta is tightly regulated. Suppressyn is a human placental endogenous retroviral protein that inhibits the profusogenic activities of another well-described endogenous retroviral protein, syncytin-1. In this study, we aimed to elucidate the mechanisms underlying suppressyn's placenta-specific expression. We identified the promoter region and a novel enhancer region for the gene encoding suppressyn, ERVH48-1, and examined their regulation via DNA methylation and their responses to changes in the oxygen concentration. Like other endogenous retroviral genes, the ERVH48-1 promoter sequence is found within a characteristic retroviral 5' LTR sequence. The novel enhancer sequence we describe here is downstream of this LTR sequence (designated EIEs: ERV internal enhancer sequence) and governs placental expression. The placenta-specific expression of ERVH48-1 is tightly controlled by DNA methylation and further regulated by oxygen concentration-dependent, hypoxia-induced transcription factors (HIF1α and HIF2α). Our findings highlight the involvement of (1) tissue specificity through DNA methylation, (2) expression specificity through placenta-specific enhancer regions, and (3) the regulation of suppressyn expression in differing oxygen conditions by HIF1α and HIF2α. We suggest that these regulatory mechanisms are central to normal and abnormal placental development, including the development of disorders of pregnancy involving altered oxygenation, such as preeclampsia, pregnancy-induced hypertension, and fetal growth restriction.
Collapse
Affiliation(s)
- Jun Sugimoto
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Hiroshima University, Hiroshima 734-8551, Japan (Y.K.)
| | - Danny J. Schust
- Department of Obstetrics and Gynecology, Duke University, Durham, NC 27710, USA
| | - Makiko Sugimoto
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Hiroshima University, Hiroshima 734-8551, Japan (Y.K.)
| | - Yoshihiro Jinno
- Department of Molecular Biology, University of the Ryukyus, Okinawa 903-0215, Japan
| | - Yoshiki Kudo
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Hiroshima University, Hiroshima 734-8551, Japan (Y.K.)
| |
Collapse
|
26
|
Zhuang BM, Cao DD, Li TX, Liu XF, Lyu MM, Wang SD, Cui XY, Wang L, Chen XL, Lin XL, Lee CL, Chiu PCN, Yeung WSB, Yao YQ. Single-cell characterization of self-renewing primary trophoblast organoids as modeling of EVT differentiation and interactions with decidual natural killer cells. BMC Genomics 2023; 24:618. [PMID: 37853336 PMCID: PMC10583354 DOI: 10.1186/s12864-023-09690-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Extravillous trophoblast cell (EVT) differentiation and its communication with maternal decidua especially the leading immune cell type natural killer (NK) cell are critical events for placentation. However, appropriate in vitro modelling system and regulatory programs of these two events are still lacking. Recent trophoblast organoid (TO) has advanced the molecular and mechanistic research in placentation. Here, we firstly generated the self-renewing TO from human placental villous and differentiated it into EVTs (EVT-TO) for investigating the differentiation events. We then co-cultured EVT-TO with freshly isolated decidual NKs for further study of cell communication. TO modelling of EVT differentiation as well as EVT interaction with dNK might cast new aspect for placentation research. RESULTS Single-cell RNA sequencing (scRNA-seq) was applied for comprehensive characterization and molecular exploration of TOs modelling of EVT differentiation and interaction with dNKs. Multiple distinct trophoblast states and dNK subpopulations were identified, representing CTB, STB, EVT, dNK1/2/3 and dNKp. Lineage trajectory and Seurat mapping analysis identified the close resemblance of TO and EVT-TO with the human placenta characteristic. Transcription factors regulatory network analysis revealed the cell-type specific essential TFs for controlling EVT differentiation. CellphoneDB analysis predicted the ligand-receptor complexes in dNK-EVT-TO co-cultures, which relate to cytokines, immunomodulation and angiogenesis. EVT was known to affect the immune properties of dNK. Our study found out that on the other way around, dNKs could exert effects on EVT causing expression changes which are functionally important. CONCLUSION Our study documented a single-cell atlas for TO and its applications on EVT differentiation and communications with dNKs, and thus provide methodology and novel research cues for future study of human placentation.
Collapse
Affiliation(s)
- Bai-Mei Zhuang
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Medical school of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Dan-Dan Cao
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China.
| | - Tian-Xi Li
- Geneplus-Shenzhen Institute, Shenzhen, China
| | - Xiao-Feng Liu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
| | - Min-Min Lyu
- Department of Clinical-Translational and Basic Research Laboratory, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Shenzhen, Futian District, Guangdong, P.R. China
| | - Si-Dong Wang
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Medical school of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xin-Yuan Cui
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
| | - Li Wang
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xiao-Lin Chen
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Xiao-Li Lin
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Cheuk-Lun Lee
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R
| | - Philip C N Chiu
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R
| | - William S B Yeung
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China
- Department of Obstetrics and Gynecology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong S.A.R
| | - Yuan-Qing Yao
- Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1st Road, Futian District, Shenzhen, Guangdong, P.R. China.
- Medical school of Chinese People's Liberation Army, Chinese People's Liberation Army General Hospital, Beijing, China.
- Department of Obstetrics and Gynecology, The First Medical Centre, Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
27
|
Li X, Li ZH, Wang YX, Liu TH. A comprehensive review of human trophoblast fusion models: recent developments and challenges. Cell Death Discov 2023; 9:372. [PMID: 37816723 PMCID: PMC10564767 DOI: 10.1038/s41420-023-01670-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023] Open
Abstract
As an essential component of the maternal-fetal interface, the placental syncytiotrophoblast layer contributes to a successful pregnancy by secreting hormones necessary for pregnancy, transporting nutrients, mediating gas exchange, balancing immune tolerance, and resisting pathogen infection. Notably, the deficiency in mononuclear trophoblast cells fusing into multinucleated syncytiotrophoblast has been linked to adverse pregnancy outcomes, such as preeclampsia, fetal growth restriction, preterm birth, and stillbirth. Despite the availability of many models for the study of trophoblast fusion, there exists a notable disparity from the ideal model, limiting the deeper exploration into the placental development. Here, we reviewed the existing models employed for the investigation of human trophoblast fusion from several aspects, including the development history, latest progress, advantages, disadvantages, scope of application, and challenges. The literature searched covers the monolayer cell lines, primary human trophoblast, placental explants, human trophoblast stem cells, human pluripotent stem cells, three-dimensional cell spheres, organoids, and placenta-on-a-chip from 1938 to 2023. These diverse models have significantly enhanced our comprehension of placental development regulation and the underlying mechanisms of placental-related disorders. Through this review, our objective is to provide readers with a thorough understanding of the existing trophoblast fusion models, making it easier to select most suitable models to address specific experimental requirements or scientific inquiries. Establishment and application of the existing human placental trophoblast fusion models.
Collapse
Affiliation(s)
- Xia Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China
| | - Zhuo-Hang Li
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China
- Medical Laboratory Department, Traditional Chinese Medicine Hospital of Yaan, 625099, Sichuan, China
| | - Ying-Xiong Wang
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China.
| | - Tai-Hang Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China.
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China.
| |
Collapse
|
28
|
Cheung VC, Bui T, Soncin F, Bai T, Kessler JA, Parast MM, Horii M. Current Strategies of Modeling Human Trophoblast Using Human Pluripotent Stem Cells in vitro. Curr Protoc 2023; 3:e875. [PMID: 37787612 PMCID: PMC10558083 DOI: 10.1002/cpz1.875] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
We previously established a trophoblast differentiation protocol from primed human pluripotent stem cells (PSC). To induce this lineage, we use a combination of Bone Morphogenetic Protein-4 (BMP4) and the WNT inhibitor IWP2. This protocol has enabled us to obtain a pure population of trophectoderm (TE)-like cells that could subsequently be terminally differentiated into syncytiotrophoblasts (STB) and extravillous trophoblasts (EVT). However, the resulting TE-like cells could only be terminally differentiated to a variable mixture of STB and EVT, with a bias toward the STB lineage. Recently, methods have been developed for derivation and culture of self-renewing human trophoblast stem cells (TSC) from human embryos and early gestation placental tissues. These primary TSCs were further able to differentiate into either STB or EVT with high efficiency using the lineage specific differentiation protocols. Based partly on these protocols, we have developed methods for establishing self-renewing TSC-like cells from PSC, and for efficient lineage-specific terminal differentiation. Here, we describe in detail the protocols to derive and maintain PSC-TSC, from both embryonic stem cells (ESC) and patient-derived induced pluripotent stem cells (iPSC), and their subsequent terminal differentiation to STB and EVT. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Trophoblast Differentiation into TE-like Cells Basic Protocol 2: Conversion of PSC-Derived TE-like Cells to TSC Basic Protocol 3: Passaging PSC-Derived TSC in iCTB Complete Medium Basic Protocol 4: STB Differentiation from PSC-derived TSC Basic Protocol 5: EVT Differentiation from PSC-derived TSC Support Protocol 1: Geltrex-coated tissue culture plate preparation Support Protocol 2: Collagen IV-coated tissue culture plate preparation Support Protocol 3: Fibronectin-coated tissue culture plate preparation.
Collapse
Affiliation(s)
- Virginia Chu Cheung
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tony Bui
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Tao Bai
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
| | - John A. Kessler
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
29
|
Lawless L, Qin Y, Xie L, Zhang K. Trophoblast Differentiation: Mechanisms and Implications for Pregnancy Complications. Nutrients 2023; 15:3564. [PMID: 37630754 PMCID: PMC10459728 DOI: 10.3390/nu15163564] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Placental development is a tightly controlled event, in which cell expansion from the trophectoderm occurs in a spatiotemporal manner. Proper trophoblast differentiation is crucial to the vitality of this gestational organ. Obstructions to its development can lead to pregnancy complications, such as preeclampsia, fetal growth restriction, and preterm birth, posing severe health risks to both the mother and offspring. Currently, the only known treatment strategy for these complications is delivery, making it an important area of research. The aim of this review was to summarize the known information on the development and mechanistic regulation of trophoblast differentiation and highlight the similarities in these processes between the human and mouse placenta. Additionally, the known biomarkers for each cell type were compiled to aid in the analysis of sequencing technologies.
Collapse
Affiliation(s)
- Lauren Lawless
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX 77030, USA;
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Yushu Qin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Ke Zhang
- Institute of Bioscience and Technology, Texas A&M University, Houston, TX 77030, USA;
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
30
|
Kanda T, Kagami K, Iizuka T, Kasama H, Matsumoto T, Sakai Y, Suzuki T, Yamamoto M, Matsuoka A, Yamazaki R, Hattori A, Horie A, Daikoku T, Ono M, Fujiwara H. Spheroid formation induces chemokine production in trophoblast-derived Swan71 cells. Am J Reprod Immunol 2023; 90:e13752. [PMID: 37491922 DOI: 10.1111/aji.13752] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/23/2023] [Accepted: 07/04/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM In the cell column of anchoring villi, the cytotrophoblast differentiates into extravillous trophoblast (EVT) and invades the endometrium in contact with maternal immune cells. Recently, chemokines were proposed to regulate the decidual immune response. To investigate the roles of chemokines around the anchoring villi, we examined the expression profiles of chemokines in the first-trimester trophoblast-derived Swan71 cells using a three-dimensional culture model. METHOD OF STUDY The gene expressions in the spheroid-formed Swan71 cells were examined by microarray and qPCR analyses. The protein expressions were examined by immunochemical staining. The chemoattractant effects of spheroid-formed Swan71 cells were examined by migration assay using monocyte-derived THP-1 cells. RESULTS The expressions of an EVT marker, laeverin, and matrix metalloproteases, MMP2 and MMP9, were increased in the spheroid-cultured Swan71 cells. Microarray and qPCR analysis revealed that mRNA expressions of various chemokines, CCL2, CCL7, CCL20, CXCL1, CXCL2, CXCL5, CXCL6, CXCL8, and CXCL10, in the spheroid-cultured Swan71 cells were up-regulated as compared with those in the monolayer-cultured Swan71 cells. These expressions were significantly suppressed by hypoxia. Migration assay showed that culture media derived from the spheroid-formed Swan71 cells promoted THP-1 cell migration. CONCLUSION This study indicated that chemokine expressions in Swan71 cells increase under a spheroid-forming culture and the culture media have chemoattractant effects. Since three-dimensional cell assembling in the spheroid resembles the structure of the cell column, this study also suggests that chemokines play important roles in the interaction between EVT and immune cells in their early differentiation stage.
Collapse
Affiliation(s)
- Tatsuhito Kanda
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Kyosuke Kagami
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Takashi Iizuka
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Haruki Kasama
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Takeo Matsumoto
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yuya Sakai
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Takuma Suzuki
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Megumi Yamamoto
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Ayumi Matsuoka
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Rena Yamazaki
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Akira Hattori
- Department of System Chemotherapy and Molecular Sciences, Kyoto University Graduate School of Pharmaceutical Sciences, Kyoto, Japan
| | - Akihito Horie
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takiko Daikoku
- Division of Animal Disease Model, Research Center for Experimental Modeling of Human Disease, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | - Hiroshi Fujiwara
- Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
31
|
Jaremek A, Shaha S, Jeyarajah MJ, Jaju Bhattad G, Chowdhury D, Riddell M, Renaud SJ. Genome-Wide Analysis of Hypoxia-Inducible Factor Binding Reveals Targets Implicated in Impaired Human Placental Syncytiotrophoblast Formation under Low Oxygen. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:846-865. [PMID: 37028593 DOI: 10.1016/j.ajpath.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 04/08/2023]
Abstract
Preeclampsia (PE) is a common and serious complication of pregnancy with no cure except premature delivery. The root cause of PE is improper development of the placenta-the temporary organ supporting fetal growth and development. Continuous formation of the multinucleated syncytiotrophoblast (STB) layer via differentiation and fusion of cytotrophoblasts (CTBs) is vital for healthy placentation and is impaired in preeclamptic pregnancies. In PE, there is reduced/intermittent placental perfusion, likely resulting in a persistently low O2 environment. Low O2 inhibits differentiation and fusion of CTBs into STB and may thus contribute to PE pathogenesis; however, the underlying mechanisms are unknown. Because low O2 activates a transcription factor complex in cells known as the hypoxia-inducible factor (HIF), the objective of this study was to investigate whether HIF signaling inhibits STB formation by regulating genes required for this process. Culture of primary CTBs, the CTB-like cell line BeWo, and human trophoblast stem cells under low O2 reduced cell fusion and differentiation into STB. Knockdown of aryl hydrocarbon receptor nuclear translocator (a key component of the HIF complex) in BeWo cells restored syncytialization and expression of STB-associated genes under different O2 levels. Chromatin immunoprecipitation sequencing facilitated the identification of global aryl hydrocarbon receptor nuclear translocator/HIF binding sites, including several near genes implicated in STB development, such as ERVH48-1 and BHLHE40, providing new insights into mechanisms underlying pregnancy diseases linked to poor placental O2 supply.
Collapse
Affiliation(s)
- Adam Jaremek
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Sumaiyah Shaha
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
| | - Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Gargi Jaju Bhattad
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Diba Chowdhury
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Meghan Riddell
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada; Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta, Canada
| | - Stephen J Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada; Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada.
| |
Collapse
|
32
|
Lechner AC, Slack JC, Carreon CK, Quade BJ, Parra-Herran C. Placental lesions attributed to shallow implantation, excess extravillous trophoblast and decidual hypoxia: Correlation with maternal vascular malperfusion and related obstetric conditions. Placenta 2023; 139:61-67. [PMID: 37329860 DOI: 10.1016/j.placenta.2023.05.020] [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: 12/20/2022] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/19/2023]
Abstract
INTRODUCTION Maternal vascular malperfusion (MVM) is one of four main patterns of placental injury defined by the Amsterdam consensus statement and is associated with adverse fetal and maternal outcomes. Laminar decidual necrosis (DLN), extravillous trophoblast islands (ETIs), placental septa (PS), and basal plate multinucleate implantation-type trophoblasts (MNTs) are lesions attributed to decidual hypoxia, excess trophoblast, and shallow implantation, but are not included in the current MVM diagnostic criteria. We aimed to investigate the relationship between these lesions and MVM. METHODS A case-control model was used to evaluate for DLN, ETIs, PS, and MNTs. Placentas with MVM on pathologic examination (defined as ≥2 related lesions) constituted the case group, and maternal age- and GPA-status-matched placentas with less than 2 lesions constituted the control group. MVM-related obstetric morbidities were recorded, including hypertension, preeclampsia, and diabetes. These were correlated with the lesions of interest. RESULTS 200 placentas were reviewed: 100 MVM cases and 100 controls. MNTs and PS showed significant enrichment in the MVM group (p < .05). Furthermore, larger foci of MNTs (>2 mm linear extent) were significantly associated with chronic or gestational hypertension (OR = 4.10; p < .05) and preeclampsia (OR = 8.14; p < .05). DLN extent correlated with placental infarction, but DLN and ETIs (including size and number) lacked association with MVM-related clinical conditions. DISCUSSION As a marker of abnormally shallow placentation and related maternal morbidities, MNT merits inclusion within the MVM pathologic spectrum. Consistent reporting of MNTs >2 mm in size is recommended, as these lesions correlate with other MVM lesions and MVM-predisposing morbidities. Other lesions, particularly DLN and ETI, lacked such association questioning their diagnostic utility.
Collapse
Affiliation(s)
- Adam C Lechner
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States; University of Missouri, Columbia, MO, United States
| | - Jonathan C Slack
- Department of Pathology, Boston Children's Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Chrystalle Katte Carreon
- Department of Pathology, Boston Children's Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Bradley J Quade
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Carlos Parra-Herran
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
33
|
Karakis V, Jabeen M, Britt JW, Cordiner A, Mischler A, Li F, San Miguel A, Rao BM. Laminin switches terminal differentiation fate of human trophoblast stem cells under chemically defined culture conditions. J Biol Chem 2023; 299:104650. [PMID: 36972789 PMCID: PMC10176266 DOI: 10.1016/j.jbc.2023.104650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Human trophoblast stem cells (hTSCs) have emerged as a powerful tool to model early placental development in vitro. Analogous to the epithelial cytotrophoblast in the placenta, hTSCs can differentiate into cells of the extravillous trophoblast (EVT) lineage or the multinucleate syncytiotrophoblast (STB). Here we present a chemically defined culture system for STB and EVT differentiation of hTSCs. Notably, in contrast to current approaches, we neither utilize forskolin for STB formation nor transforming growth factor-beta (TGFβ) inhibitors or a passage step for EVT differentiation. Strikingly, the presence of a single additional extracellular cue-laminin-111-switched the terminal differentiation of hTSCs from STB to the EVT lineage under these conditions. In the absence of laminin-111, STB formation occurred, with cell fusion comparable to that obtained with differentiation mediated by forskolin; however, in the presence of laminin-111, hTSCs differentiated to the EVT lineage. Protein expression of nuclear hypoxia-inducible factors (HIF1α and HIF2α) was upregulated during EVT differentiation mediated by laminin-111 exposure. A heterogeneous mixture of Notch1+ EVTs in colonies and HLA-G+ single-cell EVTs were obtained without a passage step, reminiscent of heterogeneity in vivo. Further analysis showed that inhibition of TGFβ signaling affected both STB and EVT differentiation mediated by laminin-111 exposure. TGFβ inhibition during EVT differentiation resulted in decreased HLA-G expression and increased Notch1 expression. On the other hand, TGFβ inhibition prevented STB formation. The chemically defined culture system for hTSC differentiation established herein facilitates quantitative analysis of heterogeneity that arises during hTSC differentiation and will enable mechanistic studies in vitro.
Collapse
Affiliation(s)
- Victoria Karakis
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Mahe Jabeen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - John W Britt
- Department of Genetics, North Carolina State University, Raleigh, North Carolina, USA
| | - Abigail Cordiner
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Adam Mischler
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Feng Li
- Department of Pathology and Laboratory Medicine, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adriana San Miguel
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA
| | - Balaji M Rao
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, USA; Golden LEAF Biomanufacturing Training and Education Center, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
34
|
Liu Z, Liu H, Wang C, Pei J, Chu N, Peng T, Li X, Gu W, Tang Y. Identification of LncRNA-miRNA-mRNA ceRNA network in hypoxia-induced HTR-8/SVneo cells for preeclampsia. Medicine (Baltimore) 2023; 102:e33649. [PMID: 37115060 PMCID: PMC10145823 DOI: 10.1097/md.0000000000033649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/26/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Preeclampsia (PE) is a common pregnancy complication, and placental hypoxia is one of its causes. We aimed to identify the transcriptional profile and construct a long non-coding RNAs (lncRNA)-centered competing endogenous RNAs (ceRNA) network in hypoxia-induced HTR8/SVneo cells. We used datasets from the GEO database to identify important pathways in PE. We performed microarray profiling and functional analysis to identify differentially expressed long non-coding RNAs (lncRNAs), differentially expressed profiles of microRNA (miRNAs), and differentially expressed profiles of messenger RNA (mRNAs) in hypoxia-induced HTR8/SVneo cells. The candidates were validated using quantitative reverse transcription polymerase chain reaction. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes analyses were performed to understand the functional significance of differentially expressed genes. Finally, we constructed an lncRNA-centered ceRNA network. Several hub genes were validated both in placentas from PE and normal pregnancy, and in hypoxia-induced HTR8/SVneo cells. The hypoxic response pathway was involved in the pathophysiology of PE. Subsequently, we identified 536 differentially expressed profiles of lncRNAs (183 upregulated and 353 downregulated), 46 differentially expressed profiles of miRNAs (35 upregulated and 11 downregulated), and 2782 differentially expressed profiles of mRNAs (DEmRNAs) (1031 upregulated and 1751 downregulated) in hypoxia-induced HTR8/SVneo cells. Gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed potential pathways affected by these genes, such as angiogenesis, the HIF-1 signaling pathway, and the PI3K-Akt signaling pathway. The ceRNA network comprised 35 lncRNAs, 11 miRNAs, 27 mRNAs, and 2 hub lncRNAs, which might play a vital role in placental functions and PE. Our results revealed the transcriptome profile and constructed an lncRNA-centered ceRNA network in hypoxia-induced HTR8/SVneo cells, thereby providing potential therapeutic targets for PE.
Collapse
Affiliation(s)
- Zhenzhen Liu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Haiyan Liu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chengjie Wang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Jiangnan Pei
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Nan Chu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Ting Peng
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaotian Li
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Weirong Gu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yao Tang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
35
|
Abstract
Establishment of the hemochorial uterine-placental interface requires exodus of trophoblast cells from the placenta and their transformative actions on the uterus, which represent processes critical for a successful pregnancy, but are poorly understood. We examined the involvement of CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxyl-terminal domain 2 (CITED2) in rat and human trophoblast cell development. The rat and human exhibit deep hemochorial placentation. CITED2 was distinctively expressed in the junctional zone (JZ) and invasive trophoblast cells of the rat. Homozygous Cited2 gene deletion resulted in placental and fetal growth restriction. Small Cited2 null placentas were characterized by disruptions in the JZ, delays in intrauterine trophoblast cell invasion, and compromised plasticity. In the human placentation site, CITED2 was uniquely expressed in the extravillous trophoblast (EVT) cell column and importantly contributed to the development of the EVT cell lineage. We conclude that CITED2 is a conserved regulator of deep hemochorial placentation.
Collapse
|
36
|
Park JH, Lee HK. The Role of Hypoxia in Brain Tumor Immune Responses. Brain Tumor Res Treat 2023; 11:39-46. [PMID: 36762807 PMCID: PMC9911710 DOI: 10.14791/btrt.2022.0043] [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: 12/07/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 02/05/2023] Open
Abstract
Oxygen is a vital component of living cells. Low levels of oxygen in body tissues, known as hypoxia, can affect multiple cellular functions across a variety of cell types and are a hallmark of brain tumors. In the tumor microenvironment, abnormal vasculature and enhanced oxygen consumption by tumor cells induce broad hypoxia that affects not only tumor cell characteristics but also the antitumor immune system. Although some immune reactions require hypoxia, hypoxia generally negatively affects immunity. Hypoxia induces tumor cell invasion, cellular adaptations to hypoxia, and tumor cell radioresistance. In addition, hypoxia limits the efficacy of immunotherapy and hinders antitumor responses. Therefore, understanding the role of hypoxia in the brain tumor, which usually does not respond to immunotherapy alone is important for the development of effective anti-tumor therapies. In this review, we discuss recent evidence supporting the role of hypoxia in the context of brain tumors.
Collapse
Affiliation(s)
- Jang Hyun Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.
| |
Collapse
|
37
|
Matsumoto S, Okamura E, Muto M, Ema M. Similarities and differences in placental development between humans and cynomolgus monkeys. Reprod Med Biol 2023; 22:e12522. [PMID: 37377753 PMCID: PMC10292683 DOI: 10.1002/rmb2.12522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Background The placenta is an extraembryonic organ, which is essential to maintain a normal pregnancy. However, placental development in humans is poorly understood because of technical and ethical reasons. Methods We analyzed the anatomical localization of each trophoblastic subtype in the cynomolgus monkey placenta by immunohistochemistry in the early second trimester. Histological differences among the mouse, cynomolgus monkey, and human placenta were compared. The PubMed database was used to search for studies on placentation in rodents and primates. Main findings The anatomical structures and subtypes of the placenta in cynomolgus monkeys are highly similar to those in humans, with the exception of fewer interstitial extravillous trophoblasts in cynomolgus monkeys. Conclusion The cynomolgus monkey appears to be a good animal model to investigate human placentation.
Collapse
Affiliation(s)
- Shoma Matsumoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Eiichi Okamura
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Masanaga Muto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life ScienceShiga University of Medical ScienceOtsuJapan
- Institute for the Advanced Study of Human Biology (WPI‐ASHBi)Kyoto UniversityKyotoJapan
| |
Collapse
|
38
|
Covarrubias A, Aguilera-Olguín M, Carrasco-Wong I, Pardo F, Díaz-Astudillo P, Martín SS. Feto-placental Unit: From Development to Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:1-29. [PMID: 37466767 DOI: 10.1007/978-3-031-32554-0_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
The placenta is an intriguing organ that allows us to survive intrauterine life. This essential organ connects both mother and fetus and plays a crucial role in maternal and fetal well-being. This chapter presents an overview of the morphological and functional aspects of human placental development. First, we describe early human placental development and the characterization of the cell types found in the human placenta. Second, the human placenta from the second trimester to the term of gestation is reviewed, focusing on the morphology and specific pathologies that affect the placenta. Finally, we focus on the placenta's primary functions, such as oxygen and nutrient transport, and their importance for placental development.
Collapse
Affiliation(s)
- Ambart Covarrubias
- Health Sciences Faculty, Universidad San Sebastián, Concepción, Chile
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillán, Chile
| | - Macarena Aguilera-Olguín
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile
- Cellular Signalling and Differentiation Laboratory (CSDL), Medicine and Science Faculty, Universidad San Sebastián, Santiago, Chile
| | - Ivo Carrasco-Wong
- Cellular Signalling and Differentiation Laboratory (CSDL), School of Medical Technology, Medicine and Science Faculty, Universidad San Sebastián, Santiago, Chile
| | - Fabián Pardo
- Metabolic Diseases Research Laboratory, Interdisciplinary Centre of Territorial Health Research (CIISTe), Biomedical Research Center (CIB), San Felipe Campus, School of Medicine, Faculty of Medicine, Universidad de Valparaíso, San Felipe, Chile
| | - Pamela Díaz-Astudillo
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile
| | - Sebastián San Martín
- Biomedical Research Centre, School of Medicine, Universidad de Valparaíso, Viña del Mar, Chile.
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile.
| |
Collapse
|
39
|
Zhou H, Zhao C, Wang P, Yang W, Zhu H, Zhang S. Regulators involved in trophoblast syncytialization in the placenta of intrauterine growth restriction. Front Endocrinol (Lausanne) 2023; 14:1107182. [PMID: 36798658 PMCID: PMC9927020 DOI: 10.3389/fendo.2023.1107182] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Placental dysfunction refers to the insufficiency of placental perfusion and chronic hypoxia during early pregnancy, which impairs placental function and causes inadequate supply of oxygen and nutrients to the fetus, affecting fetal development and health. Fetal intrauterine growth restriction, one of the most common outcomes of pregnancy-induced hypertensions, can be caused by placental dysfunction, resulting from deficient trophoblast syncytialization, inadequate trophoblast invasion and impaired vascular remodeling. During placental development, cytotrophoblasts fuse to form a multinucleated syncytia barrier, which supplies oxygen and nutrients to meet the metabolic demands for fetal growth. A reduction in the cell fusion index and the number of nuclei in the syncytiotrophoblast are found in the placentas of pregnancies complicated by IUGR, suggesting that the occurrence of IUGR may be related to inadequate trophoblast syncytialization. During the multiple processes of trophoblasts syncytialization, specific proteins and several signaling pathways are involved in coordinating these events and regulating placental function. In addition, epigenetic modifications, cell metabolism, senescence, and autophagy are also involved. Study findings have indicated several abnormally expressed syncytialization-related proteins and signaling pathways in the placentas of pregnancies complicated by IUGR, suggesting that these elements may play a crucial role in the occurrence of IUGR. In this review, we discuss the regulators of trophoblast syncytialization and their abnormal expression in the placentas of pregnancies complicated by IUGR.
Collapse
Affiliation(s)
- Hanjing Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Chenqiong Zhao
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Peixin Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Weijie Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Haiyan Zhu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- *Correspondence: Songying Zhang, ; Haiyan Zhu,
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- *Correspondence: Songying Zhang, ; Haiyan Zhu,
| |
Collapse
|
40
|
The multifaceted role of GCM1 during trophoblast differentiation in the human placenta. Proc Natl Acad Sci U S A 2022; 119:e2203071119. [PMID: 36442132 PMCID: PMC9894182 DOI: 10.1073/pnas.2203071119] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Remodeling of the uterine vasculature by invasive extravillous trophoblasts (EVTs) is a critical aspect of human placentation. Insufficient EVT invasion can lead to severe obstetrical complications like preeclampsia, intrauterine growth restriction, and preterm birth. Glial cells missing-1 (GCM1) is a transcription factor that is crucial for proper placentation in mice, and is highly expressed in human syncytiotrophoblast (ST) and EVTs. GCM1 is classically considered a master regulator of ST formation, but little is known about its contribution to the development and function of EVTs. Therefore, in this study we test the hypothesis that GCM1 is a critical regulator of both EVT and ST development and function. We show that GCM1 is highly expressed in human trophoblast stem (TS) cells differentiated into either ST or EVTs. Knockdown of GCM1 in TS cells hindered differentiation into both ST and EVT pathways. When placed in ST media, GCM1-knockdown cells formed small, unstable clusters; when placed in EVT media, cells had altered morphology and transcript profiles resembling cells trapped in an intermediate state between CT and EVT, and invasive capacity through matrix was reduced. RNA sequencing analysis of GCM1-deficient TS cells revealed downregulation of EVT-associated genes and enrichment in transcripts related to WNT signaling, which was linked to decreased expression of the EVT master regulator ASCL2 and the WNT antagonist NOTUM. Our findings reveal an essential role of GCM1 during ST and EVT development, and suggest that GCM1 regulates differentiation of human TS cells into EVTs by inducing expression of ASCL2 and NOTUM.
Collapse
|
41
|
Kojima J, Ono M, Kuji N, Nishi H. Human Chorionic Villous Differentiation and Placental Development. Int J Mol Sci 2022; 23:8003. [PMID: 35887349 PMCID: PMC9325306 DOI: 10.3390/ijms23148003] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
In humans, the placenta provides the only fetomaternal connection and is essential for establishing a pregnancy as well as fetal well-being. Additionally, it allows maternal physiological adaptation and embryonic immunological acceptance, support, and nutrition. The placenta is derived from extra-embryonic tissues that develop rapidly and dynamically in the first weeks of pregnancy. It is primarily composed of trophoblasts that differentiate into villi, stromal cells, macrophages, and fetal endothelial cells (FEC). Placental differentiation may be closely related to perinatal diseases, including fetal growth retardation (FGR) and hypertensive disorders of pregnancy (HDP), and miscarriage. There are limited findings regarding human chorionic villous differentiation and placental development because conducting in vivo studies is extremely difficult. Placental tissue varies widely among species. Thus, experimental animal findings are difficult to apply to humans. Early villous differentiation is difficult to study due to the small tissue size; however, a detailed analysis can potentially elucidate perinatal disease causes or help develop novel therapies. Artificial induction of early villous differentiation using human embryonic stem (ES) cells/induced pluripotent stem (iPS) cells was attempted, producing normally differentiated villi that can be used for interventional/invasive research. Here, we summarized and correlated early villous differentiation findings and discussed clinical diseases.
Collapse
Affiliation(s)
| | - Masanori Ono
- Department of Obstetrics and Gynecology, Tokyo Medical University, Tokyo 160-0023, Japan; (J.K.); (N.K.); (H.N.)
| | | | | |
Collapse
|
42
|
Transforming growth factor-β signaling governs the differentiation program of extravillous trophoblasts in the developing human placenta. Proc Natl Acad Sci U S A 2022; 119:e2120667119. [PMID: 35867736 PMCID: PMC9282384 DOI: 10.1073/pnas.2120667119] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Abnormal placentation has been noticed in a variety of pregnancy complications such as miscarriage, early-onset preeclampsia, and fetal growth restriction. Defects in the developmental program of extravillous trophoblasts (EVTs), migrating from placental anchoring villi into the maternal decidua and its vessels, is thought to be an underlying cause. Yet, key regulatory mechanisms controlling commitment and differentiation of the invasive trophoblast lineage remain largely elusive. Herein, comparative gene expression analyses of HLA-G-purified EVTs, isolated from donor-matched placenta, decidua, and trophoblast organoids (TB-ORGs), revealed biological processes and signaling pathways governing EVT development. In particular, bioinformatics analyses and manipulations in different versatile trophoblast cell models unraveled transforming growth factor-β (TGF-β) signaling as a crucial pathway driving differentiation of placental EVTs into decidual EVTs, the latter showing enrichment of a secretory gene signature. Removal of Wingless signaling and subsequent activation of the TGF-β pathway were required for the formation of human leukocyte antigen-G+ (HLA-G+) EVTs in TB-ORGs that resemble in situ EVTs at the level of global gene expression. Accordingly, TGF-β-treated EVTs secreted enzymes, such as DAO and PAPPA2, which were predominantly expressed by decidual EVTs. Their genes were controlled by EVT-specific induction and genomic binding of the TGF-β downstream effector SMAD3. In summary, TGF-β signaling plays a key role in human placental development governing the differentiation program of EVTs.
Collapse
|
43
|
Arbildi P, Rodríguez-Camejo C, Perelmuter K, Bollati-Fogolín M, Sóñora C, Hernández A. Hypoxia and inflammation conditions differentially affect the expression of tissue transglutaminase spliced variants and functional properties of extravillous trophoblast cells. Am J Reprod Immunol 2022; 87:e13534. [PMID: 35263002 DOI: 10.1111/aji.13534] [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: 09/02/2021] [Revised: 02/03/2022] [Accepted: 03/02/2022] [Indexed: 11/28/2022] Open
Abstract
PROBLEM Persistent hypoxia and inflammation beyond early pregnancy are involved in a bad outcome because of defective trophoblast invasiveness. Tissue transglutaminase (TG2) coregulates several cell functions. An aberrant expression and/or transamidation activity could contribute to placental dysfunction. METHOD OF STUDY The first-trimester trophoblast cell line (Swan-71) was used to study TG2 expression and cell functions in the absence or presence of inflammatory cytokines (TNF-α, IL-1β) or chemical hypoxia (CoCl2 ). We analyzed The concentration of cytokines in the supernatant by ELISA; Cell migration by scratch assay; NF-κB activation by detection of nuclear p65 by immunofluorescence or flow cytometry using a Swan-71 NF-κB-hrGFP reporter cell line. Tissue transglutaminase expression was analyzed by immunoblot and confocal microscopy. Expression of spliced mRNA variants of tissue transglutaminase was analyzed by RT-PCR. Transamidation activity was assessed by flow cytometry using 5-(biotinamido)-pentylamine substrate. RESULTS Chemical hypoxia and TGase inhibition, but not inflammatory stimuli, decreased Swan-71 migration. IL-6 production was also decreased by chemical hypoxia, but increased by inflammation. Intracellular TGase activity was increased by all stimuli, but NF-κB activation was observed only in the presence of proinflammatory cytokines. TG2 expression was decreased by CoCl2 and TNF-α. Translocation of TG2 and p65 to nuclei was observed only with TNF-α, without colocalization. Differential relative expression of spliced variants of mRNA was observed between CoCl2 and inflammatory stimuli. CONCLUSION The observed decrease in total TG2 expression and relative increase in short variants under hypoxia conditions could contribute to impaired trophoblast invasion and impact on pregnancy outcome.
Collapse
Affiliation(s)
- Paula Arbildi
- Laboratorio de Inmunología, Facultad de Ciencias/Facultad de Química, Universidad de la República, Instituto de Higiene, Montevideo, Uruguay
| | - Claudio Rodríguez-Camejo
- Laboratorio de Inmunología, Facultad de Ciencias/Facultad de Química, Universidad de la República, Instituto de Higiene, Montevideo, Uruguay
| | - Karen Perelmuter
- Cell Biology Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | | | - Cecilia Sóñora
- Laboratorio de Inmunología, Facultad de Ciencias/Facultad de Química, Universidad de la República, Instituto de Higiene, Montevideo, Uruguay.,Escuela Universitaria de Tecnología Médica (EUTM)-Facultad de Medicina, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Ana Hernández
- Laboratorio de Inmunología, Facultad de Ciencias/Facultad de Química, Universidad de la República, Instituto de Higiene, Montevideo, Uruguay
| |
Collapse
|
44
|
Choudhury J, Pandey D, Chaturvedi PK, Gupta S. Epigenetic regulation of epithelial to mesenchymal transition: a trophoblast perspective. Mol Hum Reprod 2022; 28:6572349. [PMID: 35451485 DOI: 10.1093/molehr/gaac013] [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: 02/11/2022] [Revised: 03/31/2022] [Indexed: 11/12/2022] Open
Abstract
Epigenetic changes alter expression of genes at both pre- and post-transcriptional levels without changing their DNA sequence. Accumulating evidence suggests that such changes can modify cellular behaviour and characteristics required during development and in response to various extracellular stimuli. Trophoblast cells develop from the outermost trophectoderm layer of the blastocyst and undergo many phenotypic changes as the placenta develops. One such phenotypic change is differentiation of the epithelial natured cytotrophoblasts into the mesenchymal natured extravillous trophoblasts. The extravillous trophoblasts are primarily responsible for invading into the maternal decidua and thus establishing connection with the maternal spiral arteries. Any dysregulation of this process can have adverse effects on the pregnancy outcome. Hence, tight regulation of this epithelial-mesenchymal transition is critical for successful pregnancy. This review summarizes the recent research on the epigenetic regulation of the epithelial-mesenchymal transition occurring in the trophoblast cells during placental development. The functional significance of chemical modifications of DNA and histone, which regulate transcription, as well as non-coding RNAs, which control gene expression post-transcriptionally, is discussed in relation to trophoblast biology.
Collapse
Affiliation(s)
- Jaganmoy Choudhury
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi-, 110029, India
| | - Deepak Pandey
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi-, 110029, India
| | - Pradeep Kumar Chaturvedi
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi-, 110029, India
| | - Surabhi Gupta
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi-, 110029, India
| |
Collapse
|
45
|
Physiological Function of the Dynamic Oxygen Signaling Pathway at the Maternal-fetal Interface. J Reprod Immunol 2022; 151:103626. [DOI: 10.1016/j.jri.2022.103626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 02/21/2022] [Accepted: 04/13/2022] [Indexed: 11/21/2022]
|
46
|
Shukla V, Soares MJ. Modeling Trophoblast Cell-Guided Uterine Spiral Artery Transformation in the Rat. Int J Mol Sci 2022; 23:ijms23062947. [PMID: 35328368 PMCID: PMC8950824 DOI: 10.3390/ijms23062947] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/20/2022] Open
Abstract
The rat possesses hemochorial placentation with deep intrauterine trophoblast cell invasion and trophoblast-guided uterine spiral artery remodeling, which resembles human placentation. Uterine spiral arteries are extensively remodeled to deliver sufficient supply of maternal blood and nutrients to the developing fetus. Inadequacies in these key processes negatively impact fetal growth and development. Recent innovations in genome editing combined with effective phenotyping strategies have provided new insights into placental development. Application of these research approaches has highlighted both conserved and species-specific features of hemochorial placentation. The review provides foundational information on rat hemochorial placental development and function during physiological and pathological states, especially as related to the invasive trophoblast cell-guided transformation of uterine spiral arteries. Our goal is to showcase the utility of the rat as a model for in vivo mechanistic investigations targeting regulatory events within the uterine-placental interface.
Collapse
Affiliation(s)
- Vinay Shukla
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Correspondence: (V.S.); (M.J.S.)
| | - Michael J. Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy, Kansas City, MO 64108, USA
- Correspondence: (V.S.); (M.J.S.)
| |
Collapse
|
47
|
The effects of oil sands process-affected water naphthenic acid fraction components on GDF15 secretion in extravillous trophoblast cells. Toxicol Appl Pharmacol 2022; 441:115970. [DOI: 10.1016/j.taap.2022.115970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/23/2022] [Accepted: 03/01/2022] [Indexed: 11/21/2022]
|
48
|
Park JH, Lee HK. Current Understanding of Hypoxia in Glioblastoma Multiforme and Its Response to Immunotherapy. Cancers (Basel) 2022; 14:1176. [PMID: 35267480 PMCID: PMC8909860 DOI: 10.3390/cancers14051176] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/20/2022] [Accepted: 02/23/2022] [Indexed: 02/06/2023] Open
Abstract
Hypoxia is a hallmark of glioblastoma multiforme (GBM), the most aggressive cancer of the central nervous system, and is associated with multiple aspects of tumor pathogenesis. For example, hypoxia induces resistance to conventional cancer therapies and inhibits antitumor immune responses. Thus, targeting hypoxia is an attractive strategy for GBM therapy. However, traditional studies on hypoxia have largely excluded the immune system. Recently, the critical role of the immune system in the defense against multiple tumors has become apparent, leading to the development of effective immunotherapies targeting numerous cancer types. Critically, however, GBM is classified as a "cold tumor" due to poor immune responses. Thus, to improve GBM responsiveness against immunotherapies, an improved understanding of both immune function in GBM and the role of hypoxia in mediating immune responses within the GBM microenvironment is needed. In this review, we discuss the role of hypoxia in GBM from a clinical, pathological, and immunological perspective.
Collapse
Affiliation(s)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea;
| |
Collapse
|
49
|
Iqbal K, Pierce SH, Kozai K, Dhakal P, Scott RL, Roby KF, Vyhlidal CA, Soares MJ. Evaluation of Placentation and the Role of the Aryl Hydrocarbon Receptor Pathway in a Rat Model of Dioxin Exposure. ENVIRONMENTAL HEALTH PERSPECTIVES 2021; 129:117001. [PMID: 34747641 PMCID: PMC8574979 DOI: 10.1289/ehp9256] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
BACKGROUND Our environment is replete with chemicals that can affect embryonic and extraembryonic development. Dioxins, such as 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are compounds affecting development through the aryl hydrocarbon receptor (AHR). OBJECTIVES The purpose of this investigation was to examine the effects of TCDD exposure on pregnancy and placentation and to evaluate roles for AHR and cytochrome P450 1A1 (CYP1A1) in TCDD action. METHODS Actions of TCDD were examined in wild-type and genome-edited rat models. Placenta phenotyping was assessed using morphological, biochemical, and molecular analyses. RESULTS TCDD exposures were shown to result in placental adaptations and at higher doses, pregnancy termination. Deep intrauterine endovascular trophoblast cell invasion was a prominent placentation site adaptation to TCDD. TCDD-mediated placental adaptations were dependent upon maternal AHR signaling but not upon placental or fetal AHR signaling nor the presence of a prominent AHR target, CYP1A1. At the placentation site, TCDD activated AHR signaling within endothelial cells but not trophoblast cells. Immune and trophoblast cell behaviors at the uterine-placental interface were guided by the actions of TCDD on endothelial cells. DISCUSSION We identified an AHR regulatory pathway in rats activated by dioxin affecting uterine and trophoblast cell dynamics and the formation of the hemochorial placenta. https://doi.org/10.1289/EHP9256.
Collapse
Affiliation(s)
- Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Stephen H. Pierce
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Pramod Dhakal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Regan L. Scott
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
| | - Katherine F. Roby
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Anatomy and Cell Biology, KUMC, Kansas City, Kansas, USA
| | - Carrie A. Vyhlidal
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy Kansas City, Kansas City, Missouri
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Michael J. Soares
- Institute for Reproduction and Perinatal Research, University of Kansas Medical Center (KUMC), Kansas City, Kansas, USA
- Department of Pathology and Laboratory Medicine, KUMC, Kansas City, Kansas, USA
- Center for Perinatal Research, Children’s Mercy Research Institute, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Obstetrics and Gynecology, KUMC, Kansas City, Kansas, USA
| |
Collapse
|
50
|
Sun J, Song B, Ban Y, Ma C, Sun J, Ai D, Nan Z, Wang L, Qu X. Whole transcriptome analysis of trophoblasts under hypoxia. Placenta 2021; 117:13-20. [PMID: 34768163 DOI: 10.1016/j.placenta.2021.10.007] [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: 05/27/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION A physiological hypoxia environment exists at maternal-fetal interface during early pregnancy. In addition, there is a pathological hypoxic microenvironment in patients with preeclampsia. Therefore, investigating the hypoxic adaptation and the effects of hypoxia on trophoblasts transcriptome is helpful to better understand the function and regulatory mechanism of trophoblasts at the maternal-fetal interface. METHODS Trophoblast cell line HTR-8/SVneo was cultured under normoxia and hypoxia for 24 h, the full transcriptome was analyzed via RNA-Seq. GO and KEGG enrichment were performed on differentially expressed mRNA, adjacent genes of differentially expressed lncRNA, host genes of differentially expressed circRNA and target genes of differential expressed miRNA. RESULTS The results showed that hypoxia differentially regulated 373 mRNAs, 334 lncRNAs, 71 circRNAs and 33 miRNAs. GO and KEGG enrichment showed that hypoxia negatively regulated TLR3 and PI3K-Akt signaling pathways. Consistently, we found hypoxia significantly inhibited TLR3 agonist-induced cytokines expression and the phosphorylation of Akt and mTOR. DISCUSSION Our study obtained the full transcriptome data and potential regulatory network of trophoblasts under hypoxia, providing supportive data for revealing the function of trophoblasts.
Collapse
Affiliation(s)
- Jintang Sun
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China.
| | - Bingfeng Song
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Yanli Ban
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Chao Ma
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Jia Sun
- Department of Neurology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Dan Ai
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zhaodi Nan
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Lijie Wang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xun Qu
- Laboratory of Basic Medical Sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China.
| |
Collapse
|