1
|
Zheng S, Feng W, Sun Z, Xu P, Dong S, Pan L, Shen H, He J, Chen P, Shu C. HSD17B1-mediated trophoblast differentiation lowers estrogen levels in early-onset preeclampsia. Sci Rep 2025; 15:17448. [PMID: 40394177 PMCID: PMC12092795 DOI: 10.1038/s41598-025-02490-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 05/13/2025] [Indexed: 05/22/2025] Open
Abstract
Early-onset preeclampsia (EOPE) with fetal growth restriction (FGR) is a severe hypertensive disorder of pregnancy characterized by placental dysfunction and estrogen deficiency. Based on single-cell RNA sequencing (scRNA-seq) profiling of specific placental trophoblast subtypes from EOPE-FGR and normotensive pregnancies, we identified HSD17B1, which encodes a key enzyme mediating estradiol conversion, as the central dysregulated node in EOPE pathogenesis. Multi-modal computational analysis (cluster annotation, cellular proportion calculation, comparison of differentially expressed genes, and characterization of cellular developmental trajectories) revealed key expression dynamics during syncytiotrophoblast (SCT) differentiation, with substantial suppression in EOPE specimens. Further validation using clinical placental samples confirmed the downregulation of HSD17B1 at the protein level in patients with EOPE, as demonstrated by immunohistochemistry and western blotting. Mechanistically, HSD17B1 knockdown in BeWo trophoblast models recapitulated the core EOPE phenotypes of impaired SCT differentiation and estrogen biosynthesis blockade. These findings reveal that HSD17B1 is a master coordinator of trophoblast-endocrine crosstalk, the impairment of which in placental trophoblasts may contribute to EOPE pathogenesis. Our findings provide a mechanistic basis for developing HSD17B1-targeted interventions that could contribute to the concurrent restoration of placental competence and hormonal regulation, improving the perinatal outcomes of patients with EOPE.
Collapse
Affiliation(s)
- Shu Zheng
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Wei Feng
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zewen Sun
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Peng Xu
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Shuai Dong
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Lin Pan
- The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Huimin Shen
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Jin He
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China
| | - Peng Chen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China.
| | - Chang Shu
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, Jilin University, Changchun, 130021, China.
| |
Collapse
|
2
|
Trecourt A, Donzel M, Gaillot-Durand L, Bolze PA, Golfier F, Descargues P, Hajri T, Mauduit C, Devouassoux-Shisheboran M, Allias F. SALL4 as a Useful Marker for the Distinction of Various Gestational Trophoblastic Disease Subtypes: Choriocarcinoma From Other Trophoblastic Lesions and Early Complete Hydatidiform Mole From Partial Mole and NonMolar Villi. Am J Surg Pathol 2025; 49:417-428. [PMID: 39876093 PMCID: PMC11984545 DOI: 10.1097/pas.0000000000002358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
The distinction between choriocarcinoma and residual trophoblastic cell proliferation from a complete hydatidiform mole/invasive mole (CHM/IM) without villi is challenging on curettage materials. We investigated whether SALL4 immunostaining could help differentiate various gestational trophoblastic diseases. Placental site nodules (PSN; n=10), atypical PSN (APSN; n=8), placental site trophoblastic tumors (PSTT; n=9), epithelioid trophoblastic tumors (ETT; n=5), gestational choriocarcinomas (n=31), partial hydatidiform moles (PHM; n=13), CHM/IM (n=47), and nonmolar products of conception (POC) (n=26) were included. SALL4 immunostaining was quantified (0 [1% to 10%], [11% to 100%]) and characterized (scattered single-cell or clustered nuclear positivity) in 2 locations: cytotrophoblast/intermediate trophoblast and villous stromal fibroblasts. A diffuse (11% to 100%) and clustered pattern of SALL4 immunostaining in cytotrophoblast/intermediate trophoblast was statistically associated with choriocarcinomas (74.2%, 23/31) as compared with PSN (0/10; P <0.0001), APSN (0/8; P =0.0002), PSTT (0/9; P <0.0001), ETT (0/5; P =0.0034), PHM (0/13; P <0.0001), CHM/IM (0/47; P <0.0001), and nonmolar POC (0/26; P <0.0001). Most nonchoriocarcinoma samples showed no SALL4 expression; when present, it was of low level (1% to 10%) and with a scattered single-cell staining in 3/9 PSTT (33%), 1/13 PHM (7.7%), 19/47 CHM/IM (40%), and 1/26 nonmolar POC (1.7%). These results were confirmed using a validation cohort. In addition, 66% (31/47) of CHM/IM villous stromal fibroblasts showed SALL4 expression (11% to 100%) (all before 14 gestational weeks), whereas this level of expression was never observed in PHM (0/13), nor in nonmolar POC (0/26; P <0.0001). Finally, a clustered and >10% SALL4 immunostaining in cytotrophoblast/intermediate trophoblast favors choriocarcinoma diagnosis. SALL4 expression in >10% villous stromal fibroblasts before 14 gestational weeks favors CHM/IM rather than PHM and nonmolar POC.
Collapse
Affiliation(s)
- Alexis Trecourt
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre pour l’innovation en cancérologie de Lyon (CICLY), Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon-1
| | - Marie Donzel
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1111, Centre National de la Recherche Scientifique (CNRS), Université Claude Bernard Lyon-1, Centre International de Recherche en Infectiologie (CIRI), UMR5308, Ecole Normale Supérieure de Lyon
| | - Lucie Gaillot-Durand
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre Français de Référence des maladies trophoblastique
| | - Pierre A. Bolze
- Service de Chirurgie Gynécologique et Oncologique, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Université Lyon 1, Obstétrique, CICLY, Pierre Bénite
- Centre Français de Référence des maladies trophoblastique
| | - François Golfier
- Service de Chirurgie Gynécologique et Oncologique, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Université Lyon 1, Obstétrique, CICLY, Pierre Bénite
- Centre Français de Référence des maladies trophoblastique
| | - Pierre Descargues
- Service de Chirurgie Gynécologique et Oncologique, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Université Lyon 1, Obstétrique, CICLY, Pierre Bénite
- Centre Français de Référence des maladies trophoblastique
| | - Touria Hajri
- Centre Français de Référence des maladies trophoblastique
| | - Claire Mauduit
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre Méditerranéen de Médecine Moléculaire (C3M), Institut National de la Santé et de la Recherche Médicale, Unité 1065, Nice, France
| | - Mojgan Devouassoux-Shisheboran
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre pour l’innovation en cancérologie de Lyon (CICLY), Faculté de Médecine Lyon Sud, Université Claude Bernard Lyon-1
- Centre Français de Référence des maladies trophoblastique
- Faculté de Médecine Lyon Est, Université Claude Bernard Lyon-1, Lyon
| | - Fabienne Allias
- Service de Pathologie Multi-Site, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon
- Centre Français de Référence des maladies trophoblastique
| |
Collapse
|
3
|
Lin W, Li X, An X, Yin J, Zhao Z, Yan X, Xu R, Zhao B, Du X, Cheng H, Yu Z, Yang Y. Cadmium exposure causes trophoblast abnormal syncytization and endocrine dysfunction in preeclampsia. Reprod Toxicol 2025; 135:108934. [PMID: 40300673 DOI: 10.1016/j.reprotox.2025.108934] [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: 02/11/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/01/2025]
Abstract
INTRODUCTION Cadmium exposure increases the risk of preeclampsia. Present study aimed to explore the mechanism of abnormal syncytization of trophoblasts caused by cadmium exposure and its relationship with preeclampsia. METHODS RT-PCR, Western blot and Hematoxylin-Eosin were used to detect syncytization in placenta. BeWo cells were treated with forskolin (50 μM) and CdCl2 (20 μM) to verify the mechanism through which cadmium exposure led to abnormal syncytization and cadmium exposure subsequently caused endocrine dysfunction of syncytiotrophoblast. RESULTS Compared with normotension control, abnormal accumulation of syncytial knots was found in placenta of preeclampsia. Cell fusion rate was increased and gene expressions of GCM1, SYN-1, SYN-2, PLGF and β-hCG were elevated in FSK-treated cells. In BeWo cells co-treated with FSK and CdCl2 and in BeWo cells co-treated with FSK and GCM1 siRNA, cell fusion rate was decreased and gene expression of SYN-2 was reduced as compared with FSK-treated cells. Nuclear translocation level of GCM1 was lower in placenta of preeclampsia and BeWo cells co-treated with FSK and CdCl2. The mRNA level of GCM1 and β-hCG were decreased in CdCl2 treated JEG3 cells. The expression of sFlt1 was increased and the expression of PLGF was decreased in placenta of preeclampsia, CdCl2 treated and GCM1 siRNA transfected JEG3 cells. Compared with cells treated with CdCl2 and GCM1 siRNA, co-treatment with CdCl2 and GCM1 siRNA resulted in decreased expressions of β-hCG, PLGF and GCM1 and increased expression of sFlt1 in JEG3 cells. CONCLUSION The results indicated that cadmium exposure during pregnancy led to abnormal syncytization and endocrine dysfunction of trophoblasts and were related to the occurrence of preeclampsia.
Collapse
Affiliation(s)
- Weilong Lin
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xuemeng Li
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xiangyou An
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Jiancai Yin
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Ziyan Zhao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xiaorui Yan
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Rong Xu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Baojing Zhao
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Xue Du
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Huiru Cheng
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China
| | - Zhen Yu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China.
| | - Yuanyuan Yang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), No. 81 Meishan Road, Hefei, Anhui 230032, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No. 81 Meishan Road, Hefei, Anhui 230032, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, No. 81 Meishan Road, Hefei, Anhui 230032, China.
| |
Collapse
|
4
|
Duan WK, Shaha SZ, Garcia Rivas JF, Wilson BL, Patel KJ, Domingo IK, Riddell MR. Placental cytotrophoblast microvillar stabilization is required for cell-cell fusion. Development 2025; 152:dev204619. [PMID: 40213950 PMCID: PMC12045602 DOI: 10.1242/dev.204619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 03/03/2025] [Indexed: 05/03/2025]
Abstract
The placenta is an essential organ of pregnancy required for maternal-fetal transport and communication. The surface of the placenta facing the maternal blood is formed by a single giant multinucleate cell: the syncytiotrophoblast. The syncytiotrophoblast is formed and maintained via fusion of progenitor cytotrophoblasts. Cell-cell fusion is a tightly regulated process, and in non-trophoblastic cells is accompanied by stereotypical alterations in cell shape by cells that have attained fusion-competence. The most prominent feature is the formation of actin-based membrane protrusions, but whether stereotypic morphological changes occur in fusion-competent cytotrophoblasts has not been characterized. Using a human placental explant model and trophoblast organoids, we identify microvilliation as a morphological feature that is enriched prior to fusion of cytotrophoblasts. Disruption of microvilli using an inhibitor of the actin-membrane cross-linker protein ezrin blocked cytotrophoblast fusion in both models. We provide evidence that cytotrophoblast microvilli are enriched in early endosomes and a pro-fusogenic protein. Thus, we propose that the polarized assembly of microvillar domains is crucial for mediating efficient syncytiotrophoblast development.
Collapse
Affiliation(s)
- Wendy K. Duan
- Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Sumaiyah Z. Shaha
- Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Juan F. Garcia Rivas
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Bethan L. Wilson
- Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Khushali J. Patel
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Ivan K. Domingo
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Meghan R. Riddell
- Department of Physiology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| |
Collapse
|
5
|
Keuls RA, Ochsner SA, O'Neill MB, O'Day DR, Miyauchi A, Campbell KM, Lanners N, Goldstein JA, Yee C, McKenna NJ, Parchem RJ, Parchem JG. Single-nucleus transcriptional profiling of the placenta reveals the syncytiotrophoblast stress response to COVID-19. Am J Obstet Gynecol 2025; 232:S160-S175.e7. [PMID: 40253079 DOI: 10.1016/j.ajog.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND COVID-19 in pregnancy is associated with placental immune activation, inflammation, and vascular malperfusion, but its impact on syncytiotrophoblast biology and function is unclear. OBJECTIVE This study aimed to determine the effects of maternal COVID-19 on placental syncytiotrophoblasts using single-nucleus transcriptional profiling and to compare placental stress responses in COVID-19 and preeclampsia. STUDY DESIGN For transcriptional characterization of syncytiotrophoblasts, we used the single-nucleus RNA sequencing platform, single-cell combinatorial indexing RNA sequencing (sci-RNA-seq3), to profile placental villi and fetal membranes from unvaccinated patients with symptomatic COVID-19 at birth (n = 4), gestational age-matched controls (n = 4), and a case of critical COVID-19 in the second trimester with delivery at term (n = 1). Clustering of nuclei and differential gene expression analysis was performed in Seurat. Gene ontology analysis was conducted using Enrichr. High-confidence transcriptional target analysis was used to identify key transcription factor nodes governing the syncytiotrophoblast response to maternal SARS-CoV-2 infection. Bioinformatic approaches were further used to compare the COVID-19 dataset to published preeclampsia gene signatures. Tissue analysis, including immunofluorescence, was conducted to validate the transcriptional data and to compare COVID-19 and preeclampsia placental histology for an expanded cohort of placentas: controls (n = 6), asymptomatic COVID-19 (n = 3), symptomatic COVID-19 (n = 5), and preeclampsia with severe features (n = 7). RESULTS The analyzed dataset comprised 15 cell clusters and 47,889 nuclei. We identified 3 clusters of syncytiotrophoblasts representing fusing and mature nuclei with overlapping but distinct transcriptional responses to COVID-19. Bioinformatic analyses indicated that COVID-19 is associated with the following alterations in syncytiotrophoblasts: (1) endoplasmic reticulum stress and activation of stress signaling pathways, including the unfolded protein response and integrated stress response; (2) regulation of gene expression by CCAAT/enhancer-binding protein beta (CEBPB), a master transcription factor of the syncytiotrophoblast lineage; and (3) upregulation of preeclampsia-associated genes. Using complementary methods, we confirmed increased levels of stress response proteins (eg, BiP, G3BP1) in syncytiotrophoblasts, unfolded protein response signaling (spliced XBP1 mRNA), and CEBPB activation (phosphorylation) in COVID-19. Increased cytotrophoblast proliferation (Ki-67) was also detected in COVID-19, consistent with a trophoblast response to injury. Markers of stress detected in preeclampsia demonstrated similarities in the placental stress phenotype of COVID-19 and preeclampsia. CONCLUSION Maternal COVID-19 is associated with syncytiotrophoblast endoplasmic reticulum stress and activation of the syncytiotrophoblast lineage transcription factor, CEBPB. Similarities between syncytiotrophoblast stress in COVID-19 and preeclampsia provide insights into their clinical association.
Collapse
Affiliation(s)
- Rachel A Keuls
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Scott A Ochsner
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Mary B O'Neill
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Diana R O'Day
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, WA
| | - Akihiko Miyauchi
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX
| | - Kadeshia M Campbell
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Natalie Lanners
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX
| | - Jeffery A Goldstein
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Connor Yee
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX; Larry C. Gilstrap MD Center for Perinatal and Women's Health Research, The University of Texas Health Science Center at Houston, Houston, TX
| | - Neil J McKenna
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Ronald J Parchem
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX; Department of Neuroscience, Baylor College of Medicine, Houston, TX.
| | - Jacqueline G Parchem
- Department of Obstetrics, Gynecology and Reproductive Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX.
| |
Collapse
|
6
|
Tossetta G, Fantone S, Procopio AD, Pugnaloni A, Gualtieri AF, Marzioni D. Effects of mineral fibres in an in vitro placental syncytiotrophoblast model. Curr Res Toxicol 2025; 8:100231. [PMID: 40237000 PMCID: PMC11999340 DOI: 10.1016/j.crtox.2025.100231] [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: 11/20/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
It is known that mineral fibres can be found in placental tissues, but their effect is not known on these tissues. BeWo in vitro model of syncytiotrophoblast, the outer layer of maternal-foetal barrier, is necessary to understand if mineral fibres can alter placental cell turnover and consequently to influence the outcome of pregnancy. We performed in vitro experiments using chrysotile UICC (UICC), chrysotile Valmalenco (VM) and erionite (ERI) to investigate the potential cytotoxic effects of these mineral fibres on BeWo cells. We demonstrated that all fibres are toxic while only UICC fibres caused a DNA damage that the cells were not able to repair through RAD51 activity. In addition, we demonstrated that DNA replication is not altered while cyclin D1 showed a significant decrease in VM and UICC suggesting that the cell cycle is altered in G1 phase. Moreover, UICC increased active form of caspase 3 demonstrating that apoptosis can be induced in BeWo cells. We suggest that although morphological changes are not visible in BeWo cells treated with these mineral fibres, DNA damage can lead to altered placenta physiology that can be seen late when the damage at the foetal tissues has already occurred.
Collapse
Affiliation(s)
- Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | | | - Antonio Domenico Procopio
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
- Clinical Laboratory and Molecular Diagnostic, IRCCS INRCA, Ancona, Italy
| | - Armanda Pugnaloni
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | | | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
- IRCCS INRCA, Ancona, Italy
| |
Collapse
|
7
|
Dworak H, Rozmaric T, Grillari J, Ogrodnik M. Cells of all trades - on the importance of spatial positioning of senescent cells in development, healing and aging. FEBS Lett 2025:10.1002/1873-3468.70037. [PMID: 40156464 PMCID: PMC7617592 DOI: 10.1002/1873-3468.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
Biological processes are often spatially regulated, ensuring molecular and cellular events occur in their most strategically advantageous locations. Cellular senescence, marked by cell cycle arrest and hypersecretion, is recognized as an important part of physiological processes like development and healing, but it also contributes to aging and disease. However, the spatial distribution of senescent cells and its physiological and pathological impact remain unclear. Here we compile evidence on senescent cell localization in development, healing, and aging. We emphasize the significance of their spatial patterns and speculate on the effects of disrupted spatial positioning of senescence in relation to pathologies. To summarize the specific spatial functions of senescent cells, we propose to refer to them as 'barrier' and 'conductor' functions. The 'barrier' function of senescent cells, due to their altered morphology and apoptosis resistance, separates tissues and builds a border between two environments. The conductor function, with the secretion of signaling factors, influences the surrounding area and stimulates migration, differentiation, or proliferation, among other processes. Overall, this Review explores the spatial patterning of cellular senescence in biological processes, highlighting its dual roles as 'barrier' and 'conductor' functions, and examines the implications of senescent cell distribution in development, healing, aging, and disease.
Collapse
Affiliation(s)
- Helene Dworak
- Ludwig Boltzmann Institute for Traumatology. The Research Center in cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Tomaz Rozmaric
- Ludwig Boltzmann Institute for Traumatology. The Research Center in cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology. The Research Center in cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Institute of Molecular Biotechnology, BOKU University, Vienna, Muthgasse 18, Vienna, Austria
| | - Mikolaj Ogrodnik
- Ludwig Boltzmann Institute for Traumatology. The Research Center in cooperation with AUVA, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
8
|
Benouda I, Vaiman D, Miralles F. Trophoblast Fusion in Hypertensive Disorders of Pregnancy and Preeclampsia. Int J Mol Sci 2025; 26:2859. [PMID: 40243430 PMCID: PMC11988414 DOI: 10.3390/ijms26072859] [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: 01/08/2025] [Revised: 03/11/2025] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Trophoblast fusion into the multinucleated syncytiotrophoblast (SCT) appears as an inescapable feature of placentation in mammals and other viviparous species. The trophoblast cells underlying the syncytium are considered a reservoir for the restoration of the aging peripheric structure. The transition from trophoblasts to SCTs has to be tightly regulated, and could be altered by genetic anomalies or environmental exposure. The resulting defective placental function could be one of the causes of the major placental diseases, such as preeclampsia (PE) and Intra-Uterine Growth Restriction (IUGR). This review attempts to take stock of the current knowledge about fusion mechanisms and their deregulations.
Collapse
Affiliation(s)
| | - Daniel Vaiman
- Institut Cochin, U1016, INSERM, UMR8104 CNRS, Université de Paris, 24 rue du Faubourg Saint-Jacques, 75014 Paris, France; (I.B.); (F.M.)
| | | |
Collapse
|
9
|
Gehl AL, Klawitter D, Wissenbach U, Cole M, Wesely C, Löhr H, Weissgerber P, Sota A, Meyer MR, Fecher-Trost C. The proteomic landscape of trophoblasts unravels calcium-dependent syncytialization processes and beta-chorionic gonadotropin (ß-hCG) production. Reprod Biol Endocrinol 2025; 23:33. [PMID: 40038668 PMCID: PMC11877844 DOI: 10.1186/s12958-025-01362-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The syncytiotrophoblast (STB) layer of the placenta is formed by cell fusion of cytotrophoblasts, acts as a feto-maternal barrier, is required for the production of pregnancy hormones such as chorionic gonadotropin, estradiol and progesterone and is also responsible for feto-maternal mineral exchange such as calcium. Adequate mineral supply and placental hormone production are essential for the maintenance of pregnancy, and disturbances in trophoblast integrity are associated with pregnancy complications. Since knowledge about the identity and expression levels of proteins in trophoblast and syncytiotrophoblast cells is limited so far, we analyzed the proteomes of trophoblast-like and syncytiotrophoblast-like BeWo cells under different calcium conditions. The investigation of protein expression profiles in combination with hormone assays can provide a better understanding of calcium-dependent cellular processes in trophoblasts and syncytiotrophoblasts. METHODS Here, we combine human trophoblast model cell cultures, hormone assays, antibody-based detection methods and high-resolution mass spectrometry analyzes to assess changes in cellular processes during syncytialization. RESULTS We monitored the changes in protein expression profiles during forskolin induced syncytialization of trophoblast-like cells in an unbiased manner and show that the expression of numerous proteins is strongly altered. Among them are enzymes of the glucocorticoid and sex hormones synthesis pathways such as cytochrome P450 (CYP) 19A1, CYP11A1, adrenodoxin (FDX1), hydroxysteroid dehydrogenase (HSD) 11β2 and HSD17β1, whose expression is strongly induced by syncytialization. The production of beta human chorionic gonadotropin (ß-hCG), progesterone and estradiol increase during syncytialization, while the secretion and synthesis of ß-hCG and the expression of several protein syncytiotrophoblast markers show a clear calcium dependence. CONCLUSION The broad applicability of semi-quantitative proteome profiling of cytotrophoblast- and syncytiotrophoblast-like cells provides new insights into signaling processes that occur in cytotrophoblasts /syncytiotrophoblasts during pregnancy.
Collapse
Affiliation(s)
- Anna-Lena Gehl
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Daniel Klawitter
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Ulrich Wissenbach
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Marnie Cole
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Christine Wesely
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Heidi Löhr
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Petra Weissgerber
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Adela Sota
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Markus R Meyer
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Claudia Fecher-Trost
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany.
| |
Collapse
|
10
|
Gomez Castro G, Hernández R, Cristofolini A, Portiansky E, Merkis C, Badura E, Casas L, Barbeito C, Diessler M. Histological Changes in the Cat Placenta Throughout Gestation. Vet Sci 2025; 12:207. [PMID: 40266894 PMCID: PMC11945310 DOI: 10.3390/vetsci12030207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 04/25/2025] Open
Abstract
This study on cat placental organogenesis provides a detailed histological description, emphasizing the stages that have been less described. Thirty-seven gestational sacs were obtained by ovariohysterectomy, and the gestational age of the embryos/fetuses was determined based on developmental characteristics. The placentas were measured and processed by routine histological techniques. Additionally, fresh tissue from a term placenta was processed for ultrastructural analysis. An in-depth histological analysis was performed, and several morphometric variables (placental and lamellar width, placental and labyrinthine thickness, area and number of decidual cells) were recorded and related mainly to gestational age. A significant increase was observed in fetal length from 31 dpc, while placental thickness rose until 39 dpc; lamellae became abundant, parallel, longer, and narrower. Many CTB cells gradually fused into the STB; however, it progressively reduced. Medium-sized decidual cells, arranged in groups at the junctional zone, were progressively incorporated into the lamellae; there, they persisted until term, decreasing in number and becoming larger and frequently binucleated. The description of temporal modifications in lamellar, trophoblastic, and decidual features widens current knowledge on feline placental morphogenesis. In addition, these findings might be valuable for elucidating mechanisms behind placental development, which in turn affect its efficiency.
Collapse
Affiliation(s)
- Gimena Gomez Castro
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada (LHYEDEC), Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata (FCV, UNLP), La Plata 1900, Argentina; (R.H.); (M.D.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA C1425FQB, La Plata 1900, Argentina; (A.C.); (E.P.)
| | - Rocío Hernández
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada (LHYEDEC), Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata (FCV, UNLP), La Plata 1900, Argentina; (R.H.); (M.D.)
| | - Andrea Cristofolini
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA C1425FQB, La Plata 1900, Argentina; (A.C.); (E.P.)
- Área de Microscopía Electrónica, Departamento de Patología Animal, Facultad de Agronomía y Veterinaria, Universidad Nacional de Río Cuarto, Río Cuarto 5800, Argentina;
| | - Enrique Portiansky
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA C1425FQB, La Plata 1900, Argentina; (A.C.); (E.P.)
- Laboratorio de Análisis de Imágenes (LAI), FCV, UNLP, La Plata 1900, Argentina
| | - Cecilia Merkis
- Área de Microscopía Electrónica, Departamento de Patología Animal, Facultad de Agronomía y Veterinaria, Universidad Nacional de Río Cuarto, Río Cuarto 5800, Argentina;
| | - Erika Badura
- Cátedra de Enfermedades de Aves y Pilíferos, FCV, UNLP, La Plata 1900, Argentina;
| | - Luciano Casas
- Servicio de Atención Clínica Primaria de Pequeños Animales, Hospital Escuela, FCV, UNLP, La Plata 1900, Argentina;
| | - Claudio Barbeito
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada (LHYEDEC), Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata (FCV, UNLP), La Plata 1900, Argentina; (R.H.); (M.D.)
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CABA C1425FQB, La Plata 1900, Argentina; (A.C.); (E.P.)
| | - Mónica Diessler
- Laboratorio de Histología y Embriología Descriptiva, Experimental y Comparada (LHYEDEC), Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata (FCV, UNLP), La Plata 1900, Argentina; (R.H.); (M.D.)
| |
Collapse
|
11
|
Chen Z, Zheng M, Wan T, Li J, Yuan X, Qin L, Zhang L, Hou T, Liu C, Li R. Gestational exposure to nanoplastics disrupts fetal development by promoting the placental aging via ferroptosis of syncytiotrophoblast. ENVIRONMENT INTERNATIONAL 2025; 197:109361. [PMID: 40080956 DOI: 10.1016/j.envint.2025.109361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/08/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Micro(nano)plastics (MNPs), are emerging environmental pollutants that have garnered widespread attention. Epidemiological and animal studies have shown that MNPs exposure during pregnancy is associated with adverse pregnancy outcomes, such as intrauterine growth restriction (IUGR) and miscarriage. However, the underlying mechanisms remain poorly understood. In this study, we found that exposure to a high dose (1 μg·mL-1) of 100 nm polystyrene nanoparticles (NPs) from gestational day (GD) 0 to GD17 significantly decreased fetal weight and increased the number of resorptions compared to the control group. Moreover, fetal weight was significantly lower in the high-dose group than in the low-dose (0.1 μg·mL-1) group. Meanwhile, ferroptosis and senescence were observed in placentas from mice exposed to high dose of NPs. In vitro experiments using human syncytiotrophoblast (STB) cells differentiated from BeWo cells, we found that NPs caused ferroptosis and senescence in STB cells. Subsequent investigations revealed that the inhibition of the ferroptosis signaling by ferrostain-1 (Fer-1) or deferiprone (DFP) ameliorated senescence induced by NPs in human STB cells. Furthermore, alleviating placental senescence using Fer-1 significantly improves fetal weight loss caused by NPs exposure during pregnancy in mice. Taken together, our results demonstrated that NPs exposure during pregnancy activated the ferroptosis pathway in placental STB, resulting in senescence of STB, which may attribute to the NPs-induced IUGR. This study not only elucidated the mechanistic link between NPs exposure and adverse pregnancy outcomes but also highlighted the necessity for targeted interventions to protect fetal health, underscoring the broader implications for environmental and public health policy.
Collapse
Affiliation(s)
- Zhuan Chen
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mingmeng Zheng
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Teng Wan
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Li
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiangyi Yuan
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Qin
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zhang
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tong Hou
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ran Li
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
12
|
Mansilla M, Wang Y, Nie G. Culture of cryopreserved first trimester placental tissues to study syncytial renewal. Sci Rep 2025; 15:4873. [PMID: 39929934 PMCID: PMC11810994 DOI: 10.1038/s41598-025-89022-z] [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: 09/20/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
Ex vivo studies with first trimester placental tissues are crucial for understanding human placental development, and culturing placental villi in floating conditions is vital to mimic the natural setting. Moreover, being able to cryopreserve these scarce materials for ex vivo studies would open unprecedented avenues, as they are very limited to research and the need to culture freshly adds further hurdles while limiting efficient use. Here we showed that hanging drop method is a simple yet effective approach to culture placental floating villi. We also revealed that these functional units of the early-stage human placenta can be cryopreserved for culturing, and that frozen-thawed villi can regenerate the syncytial layer following denudation. We further illustrated the utility of frozen-thawed tissues to study syncytialization, while validating the importance of HtrA4 in the process which was shown previously in cell models. These represent significant new knowledge to the field of placental biology.
Collapse
Affiliation(s)
- Mary Mansilla
- Implantation and Pregnancy Research Laboratory, School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, 3083, Australia
| | - Yao Wang
- Implantation and Pregnancy Research Laboratory, School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, 3083, Australia
| | - Guiying Nie
- Implantation and Pregnancy Research Laboratory, School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC, 3083, Australia.
| |
Collapse
|
13
|
Trinh QD, Takada K, Pham NTK, Takano C, Namiki T, Ito S, Takeda Y, Okitsu S, Ushijima H, Hayakawa S, Komine-Aizawa S. Oxidative Stress Enhances Rubella Virus Infection in Immortalized Human First-Trimester Trophoblasts. Int J Mol Sci 2025; 26:1041. [PMID: 39940811 PMCID: PMC11817118 DOI: 10.3390/ijms26031041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Rubella infection (RuV) during early pregnancy is a known cause of congenital rubella syndrome (CRS). However, the mechanisms by which the virus crosses the placenta and infects the fetus are not fully understood. It has been known that various kinds of cell stresses can occur during the placenta formation. Previously, we demonstrated that low-glucose-induced endoplasmic reticulum stress could drastically enhance RuV infection in immortalized human first-trimester trophoblast cells. In this study, we investigated the roles of oxidative stress in RuV infection in these cells. Oxidative stress was induced in Swan.71 cells by culturing them in medium containing hydrogen peroxide (H2O2) in various concentrations and durations (50 µM or 100 µM for 24 h, or 150 µM for 1 h). RuV infection with a clinical strain was performed 24 h post-treatment, and capsid proteins were visualized at 24 and 48 h post-infection (hpi) using flow cytometry (FCM) and fluorescence microscopy (IF), respectively. The findings demonstrated that oxidative stress significantly enhanced RuV infection, as evidenced by FCM analysis, showing a twofold increase in infection rate, and confirmed by IF assay. Additionally, significantly increased intracellular viral replication was observed at 3 dpi. These findings suggest that oxidative stress during early pregnancy may promote the maternal-to-fetal transmission of rubella, contributing to the development of CRS.
Collapse
Affiliation(s)
- Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
- Department of Applied Molecular Chemistry, College of Industrial Technology, Nihon University, Chiba 274-0072, Japan
| | - Chika Takano
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Takahiro Namiki
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Shun Ito
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Yoshinori Takeda
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Shoko Okitsu
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan; (K.T.); (N.T.K.P.); (C.T.); (T.N.); (S.I.); (Y.T.); (S.O.); (H.U.)
| |
Collapse
|
14
|
Uhm C, Gu J, Ju W, Pizzella S, Oktay H, Peng JYC, Guariglia S, Liu Y, Zhao H, Wang Y, Menon R, Zhong N. Single-nucleus RNA sequencing reveals distinct pathophysiological trophoblast signatures in spontaneous preterm birth subtypes. Cell Biosci 2025; 15:1. [PMID: 39773245 PMCID: PMC11705668 DOI: 10.1186/s13578-024-01343-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Spontaneous preterm birth (sPTB) poses significant challenges, affecting neonatal health and neurodevelopmental outcomes worldwide. The specific effects of placental trophoblasts on the pathological development of sPTB subtypes-preterm premature rupture of fetal membranes (pPROM) and spontaneous preterm labor (sPTL)-are not fully understood, making it crucial to uncover these impacts for the development of effective therapeutic strategies. Using single-nucleus RNA sequencing, we investigated transcriptomic and cellular differences at the maternal-fetal interface in pPROM and sPTL placentas. Our findings revealed distinct trophoblast compositions with pPROM characterized predominantly by extravillous trophoblasts (EVTs), while sPTL showed an abundance of syncytiotrophoblasts (STBs). Through cell differentiation and cell-to-cell communication analyses, other distinguishing factors were also found. In pPROM, heightened inflammation, oxidative stress, and vascular dysregulation with key pathways including tumor necrosis factor signaling, matrix metalloproteinase activation, and integrin-mediated cell adhesion, highlighted an invasive EVT profile potentially driven by hypoxic conditions and immune cell recruitment. In contrast, sPTL was marked by increased smooth muscle contraction, vascular remodeling, and altered signaling dynamics involving fibroblasts, including TGF-β and WNT pathways. Our study highlights the critical need to distinguish sPTB subtypes to improve diagnostic precision and therapeutic targeting. The molecular insights gained provide a foundation for future investigations aimed at functional validation of key pathways and exploration of trophoblasts on the development of sPTB. Ultimately, these findings pave the way for more personalized and effective interventions to mitigate adverse outcomes associated with preterm birth.
Collapse
Affiliation(s)
- Cherilyn Uhm
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Jianlei Gu
- School of Public Health, Yale University, New Haven, CT, 06520-0834, USA
| | - Weina Ju
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Stephanie Pizzella
- Department of Obstetrics and Gynecology, School of Medicine, Washington University, St. Louis, MO, 63110-1010, USA
| | - Hande Oktay
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | | | - Sararose Guariglia
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Yong Liu
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA
| | - Hongyu Zhao
- School of Public Health, Yale University, New Haven, CT, 06520-0834, USA
| | - Yong Wang
- Department of Obstetrics and Gynecology, School of Medicine, Washington University, St. Louis, MO, 63110-1010, USA
| | - Ramkumar Menon
- The University of Texas Medical Branch at Galveston, Galveston, TX, 77555-0144, USA
| | - Nanbert Zhong
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, 10314, USA.
| |
Collapse
|
15
|
Ballasy N, Apantaku I, Dean W, Hemberger M. Off to a good start: The importance of the placental exchange surface - Lessons from the mouse. Dev Biol 2025; 517:248-264. [PMID: 39491740 DOI: 10.1016/j.ydbio.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/04/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
The role of the chorio-allantoic placenta as the critical nutrient- and oxygen-supplying organ to nourish the demands of the fetus has been well recognized. This function relies on the successful establishment of the placental feto-maternal exchange unit, or interhaemal barrier, across which all nutrients as well as waste products must pass to cross from the maternal to the fetal blood circulation, or vice versa, respectively. As a consequence, defects in the establishment of this elaborate interface lead to fetal growth retardation or even embryonic lethality, depending on the severity of the defect. Beyond this essential role, however, it has also emerged that the functionality of the feto-maternal interface dictates the proper development of specific embryonic organs, with tightest links observed to the formation of the heart. In this article, we build on the foundational strength of the mouse as experimental model in which the placental causality of embryonic defects can be genetically proven. We discuss in detail the formation of the interhaemal barrier that makes up the labyrinth layer of the murine placenta, including insights into drivers of its formation and the interdependence of the cell types that make up this essential interface, from in vivo and in vitro data using mouse trophoblast stem cells. We highlight mouse genetic tools that enable the elucidation of cause-effect relationships between defects driven by either the trophoblast cells of the placenta or by embryonic cell types. We specifically emphasize gene knockouts for which a placental causality of embryonic heart defects has been demonstrated. This in-depth perspective provides much-needed insights while highlighting remaining gaps in knowledge that are essential for gaining a better understanding of the multi-facetted roles of the placenta in setting us up for a healthy start in life well beyond nutritional support alone.
Collapse
Affiliation(s)
- Noura Ballasy
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Ifeoluwa Apantaku
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Wendy Dean
- Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Dept. of Cell Biology and Anatomy, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Myriam Hemberger
- Dept. of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada.
| |
Collapse
|
16
|
Lee DS, Oster LF, Son S, Fletcher DA. Cell surface crowding is a tunable biophysical barrier to cell-cell fusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628283. [PMID: 39713336 PMCID: PMC11661186 DOI: 10.1101/2024.12.12.628283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Cell-cell fusion is fundamental to developmental processes such as muscle formation, as well as to viral infections that cause pathological syncytia. An essential step in fusion is close membrane apposition, but cell membranes are crowded with proteins, glycoproteins, and glycolipids, all of which must be cleared before a fusion pore can be nucleated. Here, we find that cell surface crowding drastically reduces fusogenicity in multiple systems, independent of the method for driving fusion. We estimate that cell surface crowding presents an energetic barrier to membrane apposition on the scale of ∼ 100 k B T , greater than that of bare membrane fusion. We show that increasing cell surface crowding reduces fusion efficiency of PEG-mediated and fusogen-mediated cell-cell fusion, as well as synthetic membranes under force. Interestingly, we find that differentiating myoblasts naturally decrease cell surface crowding prior to fusion. Cell surface crowding presents an underappreciated biophysical barrier that may be tuned developmentally and could be targeted externally to control tissue-specific cell-cell fusion.
Collapse
Affiliation(s)
- Daniel S.W. Lee
- Department of Bioengineering, University of California, Berkeley, CA 94720
| | - Liya F. Oster
- Department of Bioengineering, University of California, Berkeley, CA 94720
- Graduate Group in Biophysics, Berkeley, CA 94720
| | - Sungmin Son
- Department of Bioengineering, University of California, Berkeley, CA 94720
| | - Daniel A. Fletcher
- Department of Bioengineering, University of California, Berkeley, CA 94720
- Graduate Group in Biophysics, Berkeley, CA 94720
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720
- Chan Zuckerberg Biohub, San Francisco, CA 94158
| |
Collapse
|
17
|
Parenti M, Melough MM, Lapehn S, MacDonald J, Bammler T, Firsick EJ, Choi HY, Derefinko KJ, Enquobahrie DA, Carroll KN, LeWinn KZ, Bush NR, Zhao Q, Sathyanarayana S, Paquette AG. Associations Between Prenatal Vitamin D and Placental Gene Expression. J Nutr 2024; 154:3603-3614. [PMID: 39401684 DOI: 10.1016/j.tjnut.2024.10.019] [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: 05/31/2024] [Revised: 09/16/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Vitamin D is a hormone that regulates gene transcription. Prenatal vitamin D has been linked to immune and vascular function in the placenta, a key organ of pregnancy. Transcriptome-wide RNA sequencing can provide a more complete representation of the placental effects of vitamin D. OBJECTIVES We investigated the association between prenatal vitamin D concentrations and placental gene expression in a large, prospective pregnancy cohort. METHODS Participants were recruited from Shelby County, TN, United States, in the Conditions Affecting Neurocognitive Development and Learning in Early childhood (CANDLE) study. Vitamin D (plasma total 25-hydroxyvitatmin D, [25(OH)D]) was measured at midpregnancy (16-28 wk) and delivery. RNA was sequenced from placental samples collected at birth. We identified differentially expressed genes (DEGs) using adjusted linear regression models. We also conducted weighted gene coexpression network analysis. RESULTS The median 25(OH)D of participants was 21.8 ng/mL at midpregnancy (N = 774; IQR: 15.4-26.5 ng/mL) and 23.6 ng/mL at delivery (n = 753; IQR: 16.8-29.1 ng/mL). Placental expression of 17 DEGs was associated with 25(OH)D at midpregnancy, but only 1 DEG was associated with 25(OH)D at delivery. DEGs were related to energy metabolism, cytoskeletal function, and transcriptional regulation. We identified 2 weighted gene coexpression network analysis gene modules whose expression was associated with 25(OH)D at midpregnancy and 1 module associated with 25(OH)D at delivery. These modules were enriched for genes related to mitochondrial and cytoskeletal function and were regulated by transcription factors including ARNT2 and FOSL2. We also identified 12 modules associated with 25(OH)D in females and 1 module in males. CONCLUSIONS 25(OH)D during midpregnancy, but not at delivery, is associated with placental gene expression at birth. Future research is needed to investigate a potential role of vitamin D in modulating placental mitochondrial metabolism, intracellular transport, and transcriptional regulation during pregnancy.
Collapse
Affiliation(s)
- Mariana Parenti
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States.
| | - Melissa M Melough
- Department of Health Behavior and Nutrition Sciences, University of Delaware, Newark, DE, United States
| | - Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Evan J Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States
| | - Hyo Young Choi
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United States
| | - Karen J Derefinko
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United States; Department of Pharmacology, Addiction Science, and Toxicology, University of Tennessee Health Sciences Center, Memphis, TN, United States
| | - Daniel A Enquobahrie
- Department of Epidemiology, University of Washington, Seattle, WA, United States
| | - Kecia N Carroll
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kaja Z LeWinn
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States; Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Qi Zhao
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United States
| | - Sheela Sathyanarayana
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States; Department of Epidemiology, University of Washington, Seattle, WA, United States; Center for Child Health, Behavior, and Development, Seattle Children's Research Institute, Seattle, WA, United States; Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, United States; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States; Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
18
|
Parameshwar PK, Vaillancourt C, Moraes C. Engineering placental trophoblast fusion: A potential role for biomechanics in syncytialization. Placenta 2024; 157:50-54. [PMID: 38448351 DOI: 10.1016/j.placenta.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
The process by which placental trophoblasts fuse to form the syncytiotrophoblast around the chorionic villi is not fully understood. Mechanical features of the in vivo and in vitro culture environments have recently emerged as having the potential to influence fusion efficiency, and considering these mechanical cues may ultimately allow predictive control of trophoblast syncytialization. Here, we review recent studies that suggest that biomechanical factors such as shear stress, tissue stiffness, and dimensionally-related stresses affect villous trophoblast fusion efficiency. We then discuss how these stimuli might arise in vivo and how they can be incorporated in cultures to study and enhance villous trophoblast fusion. We believe that this mechanical paradigm will provide novel insight into manipulating the syncytialization process to better engineer improved models, understand disease progression, and ultimately develop novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Cathy Vaillancourt
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, H7B 1B7, Canada; Department of Obstetrics and Gynecology, Université de Montréal, and Research Center Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) du Nord-de-l'Île-de-Montréal, Montréal, QC, H3L 1K5, Canada
| | - Christopher Moraes
- Department of Biological and Biomedical Engineering, McGill University, Montréal, QC, H3A 2B4, Canada; Department of Chemical Engineering, McGill University, Montréal, QC, H3A 0C5, Canada; Goodman Cancer Research Centre, McGill University, Montréal, QC, H3A 1A3, Canada; Division of Experimental Medicine, McGill University, Montréal, QC, H4A 3J1, Canada.
| |
Collapse
|
19
|
Baker JC. Placental Evolution: Innovating How to Feed Babies. Annu Rev Genet 2024; 58:391-408. [PMID: 39227137 DOI: 10.1146/annurev-genet-111523-102135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The evolution of the placenta was transformative. It changed how offspring are fed during gestation from depositing all the resources into an egg to continually supplying resources throughout gestation. Placental evolution is infinitely complex, with many moving parts, but at the core it is driven by a conflict over resources between the mother and the baby, which sets up a Red Queen race, fueling rapid diversification of morphological, cellular, and genetic forms. Placentas from even closely related species are highly divergent in form and function, and many cellular processes are distinct. If we could extract the entirety of genomic information for placentas across all species, including the many hundreds that have evolved in fish and reptiles, we could find their shared commonality, and that would tell us which of the many pieces really matter. We do not have this information, but we do have clues. Convergent evolution mechanisms were repeatedly used in the placenta, including the intense selective pressure to co-opt an envelope protein to build a multinucleated syncytium, the use of the same hormones and structural proteins in placentas derived from separate embryonic origins that arose hundreds of millions of years apart, and the co-option of endogenous retroviruses to form capsids as a way of transport and as mutagens to form new enhancers. As a result, the placental genome is the Wild West of biology, set up to rapidly change, adapt, and innovate. This ability to adapt facilitated the evolution of big babies with big brains and will continue to support offspring and their mothers in our ever-changing global environment.
Collapse
Affiliation(s)
- Julie C Baker
- Department of Genetics, Stanford University, Stanford, California, USA;
| |
Collapse
|
20
|
Abostait A, Abdelkarim M, Bao Z, Miyake Y, Tse WH, Di Ciano-Oliveir C, Buerki-Thurnherr T, Allen C, Keijzer R, Labouta HI. Optimizing lipid nanoparticles for fetal gene delivery in vitro, ex vivo, and aided with machine learning. J Control Release 2024; 376:678-700. [PMID: 39447842 DOI: 10.1016/j.jconrel.2024.10.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
There is a clinical need to develop lipid nanoparticles (LNPs) to deliver congenital therapies to the fetus during pregnancy. The aim of these therapies is to restore normal fetal development and prevent irreversible conditions after birth. As a first step, LNPs need to be optimized for transplacental transport, safety on the placental barrier and fetal organs and transfection efficiency. We developed and characterized a library of LNPs of varying compositions and used machine learning (ML) models to delineate the determinants of LNP size and zeta potential. Utilizing different in vitro placental models with the help of a Random Forest algorithm, we could identify the top features driving percentage LNP transport and kinetics at 24 h, out of a total of 18 input features represented by 41 LNP formulations and 48 different transport experiments. We further evaluated the LNPs for safety, placental cell uptake, transfection efficiency in placental trophoblasts and fetal lung fibroblasts. To ensure the integrity of the LNPs following transplacental transport, we screened LNPs for transport and transfection using a high-throughput integrated transport-transfection in vitro model. Finally, we assessed toxicity of the LNPs in a tracheal occlusion fetal lung explant model. LNPs showed little to no toxicity to fetal and placental cells. Immunoglobin G (IgG) orientation on the surface of LNPs, PEGylated lipids, and ionizable lipids had significant effects on placental transport. The Random Forest algorithm identified the top features driving LNPs placental transport percentage and kinetics. Zeta potential emerged in the top driving features. Building on the ML model results, we developed new LNP formulations to further optimize the transport leading to 622 % increase in transport at 24 h versus control LNP formulation. To induce preferential siRNA transfection of fetal lung, we further optimized cationic lipid percentage and the lipid-to-siRNA ratio. Studying LNPs in an integrated placental and fetal lung fibroblasts model showed a strong correlation between zeta potential and fetal lung transfection. Finally, we assessed the toxicity of LNPs in a tracheal occlusion lung explant model. The optimized formulations appeared to be safe on ex vivo fetal lungs as indicated by insignificant changes in apoptosis (Caspase-3) and proliferation (Ki67) markers. In conclusion, we have optimized an LNP formulation that is safe, with high transplacental transport and preferential transfection in fetal lung cells. Our research findings represent an important step toward establishing the safety and effectiveness of LNPs for gene delivery to the fetal organs.
Collapse
Affiliation(s)
- Amr Abostait
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada
| | - Mahmoud Abdelkarim
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto M5S 3G9, Canada
| | - Zeqing Bao
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Yuichiro Miyake
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada; Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo Bunkyo-ku, Tokyo 113-8421, Japan
| | - Wai Hei Tse
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | | | - Tina Buerki-Thurnherr
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), St. Gallen 9014, Switzerland
| | - Christine Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Richard Keijzer
- Department of Surgery, Division of Pediatric Surgery, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba and Children's Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Hagar I Labouta
- Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto M5B 1T8, Canada; College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada; Biomedical Engineering, Faculty of Engineering, University of Toronto, Toronto M5S 3G9, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada.
| |
Collapse
|
21
|
Jeyarajah MJ, Patterson VS, Jaju Bhattad G, Zhao L, Whitehead SN, Renaud SJ. Placental extracellular vesicles promote cardiomyocyte maturation and fetal heart development. Commun Biol 2024; 7:1254. [PMID: 39363116 PMCID: PMC11450004 DOI: 10.1038/s42003-024-06938-4] [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: 04/14/2023] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
Congenital heart defects are leading causes of neonatal mortality and are often associated with placental abnormalities, but mechanisms linking placenta and heart development are poorly understood. Herein, we investigated a potential signaling network connecting the placenta and nascent heart in mice. We found that fetal hearts exposed to media conditioned by placental tissue or differentiated wild-type trophoblast stem (TS) cells, but not undifferentiated TS cells, showed increased heart rate and epicardial cell outgrowth. This effect was not observed when hearts were exposed to media from TS cells lacking OVO-Like 2, a transcription factor required for trophoblast differentiation and placental development. Trophoblasts released abundant extracellular vesicles into media, and these vesicles were sufficient to mediate cardio-promoting effects. Our findings provide a potential mechanism whereby the placenta communicates with the fetal heart to promote cardiac morphogenesis, and offers insight into the link between poor placentation and a higher incidence of heart defects.
Collapse
Affiliation(s)
- Mariyan J Jeyarajah
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Violet S Patterson
- 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
| | - Lin Zhao
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Shawn N Whitehead
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, 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, London, Ontario, Canada.
| |
Collapse
|
22
|
Li X, Shao LZ, Li ZH, Wang YH, Cai QY, Wang S, Chen H, Sheng J, Luo X, Chen XM, Wang YX, Ding YB, Liu TH. STK40 inhibits trophoblast fusion by mediating COP1 ubiquitination to degrade P57 Kip2. J Transl Med 2024; 22:852. [PMID: 39304928 PMCID: PMC11414097 DOI: 10.1186/s12967-024-05360-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: 02/06/2024] [Accepted: 05/29/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The syncytiotrophoblast (SCT) layer in the placenta serves as a crucial physical barrier separating maternal-fetal circulation, facilitating essential signal and substance exchange between the mother and fetus. Any abnormalities in its formation or function can result in various maternal syndromes, such as preeclampsia. The transition of proliferative villous cytotrophoblasts (VCT) from the mitotic cell cycle to the G0 phase is a prerequisite for VCT differentiation and their fusion into SCT. The imprinting gene P57Kip2, specifically expressed in intermediate VCT capable of fusion, plays a pivotal role in driving this key event. Moreover, aberrant expression of P57Kip2 has been linked to pathological placental conditions and adverse fetal outcomes. METHODS Validation of STK40 interaction with P57Kip2 using rigid molecular simulation docking and co-immunoprecipitation. STK40 expression was modulated by lentivirus in BeWo cells, and the effect of STK40 on trophoblast fusion was assessed by real-time quantitative PCR, western blot, immunofluorescence, and cell viability and proliferation assays. Co-immunoprecipitation, transcriptome sequencing, and western blot were used to determine the potential mechanisms by which STK40 regulates P57Kip2. RESULTS In this study, STK40 has been identified as a novel interacting protein with P57Kip2, and its expression is down-regulated during the fusion process of trophoblast cells. Overexpressing STK40 inhibited cell fusion in BeWo cells while stimulating mitotic cell cycle activity. Further experiments indicated that this effect is attributed to its specific binding to the CDK-binding and the Cyclin-binding domains of P57Kip2, mediating the E3 ubiquitin ligase COP1-mediated ubiquitination and degradation of P57Kip2. Moreover, abnormally high expression of STK40 might significantly contribute to the occurrence of preeclampsia. CONCLUSIONS This study offers new insights into the role of STK40 in regulating the protein-level homeostasis of P57Kip2 during placental development.
Collapse
Affiliation(s)
- Xia Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
| | - Li-Zhen Shao
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
| | - Zhuo-Hang Li
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
- Medical Laboratory Department, Traditional Chinese Medicine Hospital of Yaan, Sichuan, 625099, China
| | - Yong-Heng Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
| | - Qin-Yu Cai
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
| | - Shun Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
| | - Jie Sheng
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
| | - Xin Luo
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, PR China
| | - Xue-Mei Chen
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China
| | - Ying-Xiong Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China.
| | - Yu-Bin Ding
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China.
| | - Tai-Hang Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400016, China.
- The Joint International Research Laboratory of Reproduction and Development, Chongqing Medical University, Box 197, No.1 Yixueyuan Rd, Chongqing, 400016, China.
| |
Collapse
|
23
|
Forndran T, Große S, Weber G, Hausdorf L, Samsel D, Berndt A, Gaßler N, Groten T. Protocol for the quantitative analysis of images retrieved by multiplex immunofluorescence staining to allow cell type-specific spatial phenotyping of markers of interest in the human placenta. Placenta 2024. [DOI: 10.1016/j.placenta.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
24
|
Zheng W, Zhang Y, Xu P, Wang Z, Shao X, Chen C, Cai H, Wang Y, Sun MA, Deng W, Liu F, Lu J, Zhang X, Cheng D, Mysorekar IU, Wang H, Wang YL, Hu X, Cao B. TFEB safeguards trophoblast syncytialization in humans and mice. Proc Natl Acad Sci U S A 2024; 121:e2404062121. [PMID: 38968109 PMCID: PMC11253012 DOI: 10.1073/pnas.2404062121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 05/17/2024] [Indexed: 07/07/2024] Open
Abstract
Nutrient sensing and adaptation in the placenta are essential for pregnancy viability and proper fetal growth. Our recent study demonstrated that the placenta adapts to nutrient insufficiency through mechanistic target of rapamycin (mTOR) inhibition-mediated trophoblast differentiation toward syncytiotrophoblasts (STBs), a highly specialized multinucleated trophoblast subtype mediating extensive maternal-fetal interactions. However, the underlying mechanism remains elusive. Here, we unravel the indispensable role of the mTORC1 downstream transcriptional factor TFEB in STB formation both in vitro and in vivo. TFEB deficiency significantly impaired STB differentiation in human trophoblasts and placenta organoids. Consistently, systemic or trophoblast-specific deletion of Tfeb compromised STB formation and placental vascular construction, leading to severe embryonic lethality. Mechanistically, TFEB conferred direct transcriptional activation of the fusogen ERVFRD-1 in human trophoblasts and thereby promoted STB formation, independent of its canonical function as a master regulator of the autophagy-lysosomal pathway. Moreover, we demonstrated that TFEB directed the trophoblast syncytialization response driven by mTOR complex 1 (mTORC1) signaling. TFEB expression positively correlated with the reinforced trophoblast syncytialization in human fetal growth-restricted placentas exhibiting suppressed mTORC1 activity. Our findings substantiate that the TFEB-fusogen axis ensures proper STB formation during placenta development and under nutrient stress, shedding light on TFEB as a mechanistic link between nutrient-sensing machinery and trophoblast differentiation.
Collapse
Affiliation(s)
- Wanshan Zheng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Yue Zhang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Peiqun Xu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Zexin Wang
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen361102, Fujian, China
| | - Xuan Shao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing100101, China
- University of Chinese Academy of Sciences, Beijing100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing100101, China
| | - Chunyan Chen
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Yinan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Ming-an Sun
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou225009, Jiangsu, China
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Fan Liu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Xueqin Zhang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Dunjin Cheng
- Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou510140, Guangdong, China
- Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou510140, Guangdong, China
| | - Indira U. Mysorekar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston77030, TX
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston77030, TX
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing100101, China
- University of Chinese Academy of Sciences, Beijing100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing100101, China
| | - Xiaoqian Hu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, Xiamen361102, Fujian, China
| | - Bin Cao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| |
Collapse
|
25
|
Shimada H, Powell TL, Jansson T. Regulation of placental amino acid transport in health and disease. Acta Physiol (Oxf) 2024; 240:e14157. [PMID: 38711335 PMCID: PMC11162343 DOI: 10.1111/apha.14157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
Abnormal fetal growth, i.e., intrauterine growth restriction (IUGR) or fetal growth restriction (FGR) and fetal overgrowth, is associated with increased perinatal morbidity and mortality and is strongly linked to the development of metabolic and cardiovascular disease in childhood and later in life. Emerging evidence suggests that changes in placental amino acid transport may contribute to abnormal fetal growth. This review is focused on amino acid transport in the human placenta, however, relevant animal models will be discussed to add mechanistic insights. At least 25 distinct amino acid transporters with different characteristics and substrate preferences have been identified in the human placenta. Of these, System A, transporting neutral nonessential amino acids, and System L, mediating the transport of essential amino acids, have been studied in some detail. Importantly, decreased placental Systems A and L transporter activity is strongly associated with IUGR and increased placental activity of these two amino acid transporters has been linked to fetal overgrowth in human pregnancy. An array of factors in the maternal circulation, including insulin, IGF-1, and adiponectin, and placental signaling pathways such as mTOR, have been identified as key regulators of placental Systems A and L. Studies using trophoblast-specific gene targeting in mice have provided compelling evidence that changes in placental Systems A and L are mechanistically linked to altered fetal growth. It is possible that targeting specific placental amino acid transporters or their upstream regulators represents a novel intervention to alleviate the short- and long-term consequences of abnormal fetal growth in the future.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
- Departments of Obstetrics & Gynecology, Sapporo Medical University, Sapporo, Japan
| | - Theresa L Powell
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO, US
| | - Thomas Jansson
- Department of Obstetrics and Gynecology University of Colorado, Anschutz Medical Campus, Aurora, CO, US
| |
Collapse
|
26
|
Kotta-Loizou I, Pritsa A, Antasouras G, Vasilopoulos SN, Voulgaridou G, Papadopoulou SK, Coutts RHA, Lechouritis E, Giaginis C. Fetus Exposure to Drugs and Chemicals: A Holistic Overview on the Assessment of Their Transport and Metabolism across the Human Placental Barrier. Diseases 2024; 12:114. [PMID: 38920546 PMCID: PMC11202568 DOI: 10.3390/diseases12060114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/23/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND The placenta exerts a crucial role in fetus growth and development during gestation, protecting the fetus from maternal drugs and chemical exposure. However, diverse drugs and chemicals (xenobiotics) can penetrate the maternal placental barrier, leading to deleterious, adverse effects concerning fetus health. Moreover, placental enzymes can metabolize drugs and chemicals into more toxic compounds for the fetus. Thus, evaluating the molecular mechanisms through which drugs and chemicals transfer and undergo metabolism across the placental barrier is of vital importance. In this aspect, this comprehensive literature review aims to provide a holistic approach by critically summarizing and scrutinizing the potential molecular processes and mechanisms governing drugs and chemical transfer and metabolism across the placental barrier, which may lead to fetotoxicity effects, as well as analyzing the currently available experimental methodologies used to assess xenobiotics placental transfer and metabolism. METHODS A comprehensive and in-depth literature review was conducted in the most accurate scientific databases such as PubMed, Scopus, and Web of Science by using relevant and effective keywords related to xenobiotic placental transfer and metabolism, retrieving 8830 published articles until 5 February 2024. After applying several strict exclusion and inclusion criteria, a final number of 148 relevant published articles were included. RESULTS During pregnancy, several drugs and chemicals can be transferred from the mother to the fetus across the placental barrier by either passive diffusion or through placental transporters, resulting in fetus exposure and potential fetotoxicity effects. Some drugs and chemicals also appear to be metabolized across the placental barrier, leading to more toxic products for both the mother and the fetus. At present, there is increasing research development of diverse experimental methodologies to determine the potential molecular processes and mechanisms of drug and chemical placental transfer and metabolism. All the currently available methodologies have specific strengths and limitations, highlighting the strong demand to utilize an efficient combination of them to obtain reliable evidence concerning drug and chemical transfer and metabolism across the placental barrier. To derive the most consistent and safe evidence, in vitro studies, ex vivo perfusion methods, and in vivo animal and human studies can be applied together with the final aim to minimize potential fetotoxicity effects. CONCLUSIONS Research is being increasingly carried out to obtain an accurate and safe evaluation of drug and chemical transport and metabolism across the placental barrier, applying a combination of advanced techniques to avoid potential fetotoxic effects. The improvement of the currently available techniques and the development of novel experimental protocols and methodologies are of major importance to protect both the mother and the fetus from xenobiotic exposure, as well as to minimize potential fetotoxicity effects.
Collapse
Affiliation(s)
- Ioly Kotta-Loizou
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK;
| | - Agathi Pritsa
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Georgios Antasouras
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| | - Spyridon N. Vasilopoulos
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou Campus, 15780 Athens, Greece;
| | - Gavriela Voulgaridou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Sousana K. Papadopoulou
- Department of Nutritional Sciences and Dietetics, School of Health Sciences, International Hellenic University, 57400 Thessaloniki, Greece; (G.V.); (S.K.P.)
| | - Robert H. A. Coutts
- Department of Clinical, Pharmaceutical and Biological Sciences, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9AB, UK;
| | - Eleftherios Lechouritis
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| | - Constantinos Giaginis
- Department of Food Science and Nutrition, School of the Environment, University of the Aegean, 81400 Lemnos, Greece; (G.A.); (E.L.); (C.G.)
| |
Collapse
|
27
|
Parameshwar PK, Li C, Arnauts K, Jiang J, Rostami S, Campbell BE, Lu H, Rosenzweig DH, Vaillancourt C, Moraes C. Directed biomechanical compressive forces enhance fusion efficiency in model placental trophoblast cultures. Sci Rep 2024; 14:11312. [PMID: 38760496 PMCID: PMC11101427 DOI: 10.1038/s41598-024-61747-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/09/2024] [Indexed: 05/19/2024] Open
Abstract
The syncytiotrophoblast is a multinucleated structure that arises from fusion of mononucleated cytotrophoblasts, to sheath the placental villi and regulate transport across the maternal-fetal interface. Here, we ask whether the dynamic mechanical forces that must arise during villous development might influence fusion, and explore this question using in vitro choriocarcinoma trophoblast models. We demonstrate that mechanical stress patterns arise around sites of localized fusion in cell monolayers, in patterns that match computational predictions of villous morphogenesis. We then externally apply these mechanical stress patterns to cell monolayers and demonstrate that equibiaxial compressive stresses (but not uniaxial or equibiaxial tensile stresses) enhance expression of the syndecan-1 and loss of E-cadherin as markers of fusion. These findings suggest that the mechanical stresses that contribute towards sculpting the placental villi may also impact fusion in the developing tissue. We then extend this concept towards 3D cultures and demonstrate that fusion can be enhanced by applying low isometric compressive stresses to spheroid models, even in the absence of an inducing agent. These results indicate that mechanical stimulation is a potent activator of cellular fusion, suggesting novel avenues to improve experimental reproductive modelling, placental tissue engineering, and understanding disorders of pregnancy development.
Collapse
Affiliation(s)
| | - Chen Li
- Department of Chemical Engineering, McGill University, Montréal, Québec, Canada
| | - Kaline Arnauts
- Department of Chemical Engineering, McGill University, Montréal, Québec, Canada
| | - Junqing Jiang
- Department of Chemical Engineering, McGill University, Montréal, Québec, Canada
| | - Sabra Rostami
- Department of Chemical Engineering, McGill University, Montréal, Québec, Canada
| | - Benjamin E Campbell
- Department of Chemical Engineering, McGill University, Montréal, Québec, Canada
| | - Hongyan Lu
- Department of Chemical Engineering, McGill University, Montréal, Québec, Canada
| | - Derek Hadar Rosenzweig
- Department of Surgery, McGill University, Montréal, Québec, Canada
- Injury, Repair and Recovery Program, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Cathy Vaillancourt
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, Québec, Canada
- Department of Obstetrics and Gynecology, Université de Montréal, and Research Center Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) du Nord-de-L'Île-de-Montréal, Montréal, Québec, Canada
| | - Christopher Moraes
- Department of Biological and Biomedical Engineering, McGill University, Montréal, Québec, Canada.
- Department of Chemical Engineering, McGill University, Montréal, Québec, Canada.
- Goodman Cancer Research Centre, McGill University, Montréal, Québec, Canada.
- Division of Experimental Medicine, McGill University, Montréal, Québec, Canada.
| |
Collapse
|
28
|
Slaby EM, Hansen N, Sharma R, Pirrotte P, Weaver JD. Engineered 3D Hydrogel Matrices to Modulate Trophoblast Stem Cell-Derived Placental Organoid Phenotype. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.13.594007. [PMID: 38798435 PMCID: PMC11118344 DOI: 10.1101/2024.05.13.594007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Placental organoid models are a promising platform to study human placental development and function. Organoid systems typically use naturally derived hydrogel extracellular matrices (ECM), resulting in batch-to-batch variability that limits experimental reproducibility. As an alternative, synthetic ECM-mimicking hydrogel matrices offer greater consistency and control over environmental cues. Here, we generated trophoblast stem cell-derived placental organoids using poly(ethylene glycol) (PEG) hydrogels with tunable degradability and placenta-derived ECM cues to evaluate trophoblast differentiation relative to Matrigel and two-dimensional (2D) culture controls. Our data demonstrate that PEG hydrogels support trophoblast viability and metabolic function comparable to gold standard Matrigel. Additionally, phenotypic characterization via proteomic analysis revealed that PEG and Matrigel matrices drive syncytiotrophoblast and extravillous trophoblast-dominant placental organoid phenotypes, respectively. Further, three-dimensional (3D) environments promoted greater integrin expression and ECM production than 2D culture. This study demonstrates that engineered 3D culture environments can be used to reliably generate placental organoids and guide trophoblast differentiation.
Collapse
|
29
|
Parenti M, Melough MM, Lapehn S, MacDonald J, Bammler T, Firsick EJ, Choi HY, Derefinko KJ, Enquobahrie DA, Carroll KN, LeWinn KZ, Bush NR, Zhao Q, Sathyanarayana S, Paquette AG. Associations Between Prenatal Vitamin D and Placental Gene Expression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593571. [PMID: 38765981 PMCID: PMC11100832 DOI: 10.1101/2024.05.10.593571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background Vitamin D is a hormone regulating gene transcription. Prenatal vitamin D has been linked to immune and vascular function in the placenta, a key organ of pregnancy. To date, studies of vitamin D and placental gene expression have focused on a limited number of candidate genes. Transcriptome-wide RNA sequencing can provide a more complete representation of the placental effects of vitamin D. Objective We investigated the association between prenatal vitamin D levels and placental gene expression in a large, prospective pregnancy cohort. Methods Participants were recruited in Shelby County, Tennessee in the Conditions Affecting Neurocognitive Development and Learning in Early childhood (CANDLE) study. Vitamin D level (plasma total 25-hydroxyvitatmin D, [25(OH)D]) was measured at mid-pregnancy (16-28 weeks' gestation) and delivery. Placenta samples were collected at birth. RNA was isolated and sequenced. We identified differentially expressed genes (DEGs) using adjusted linear regression models. We also conducted weighted gene co-expression network analysis (WGCNA). Results The median 25(OH)D of participants was 21.8 ng/mL at mid-pregnancy (N=774, IQR: 15.4-26.5 ng/mL) and 23.6 ng/mL at delivery (N=753, IQR: 16.8-29.1 ng/mL). Placental expression of 25 DEGs was associated with 25(OH)D at mid-pregnancy, but no DEG was associated with 25(OH)D at delivery. DEGs were related to energy metabolism, cytoskeletal function, and RNA transcription. Using WGCNA, we identified 2 gene modules whose expression was associated with 25(OH)D at mid-pregnancy and 1 module associated with 25(OH)D at delivery. These modules were enriched for genes related to mitochondrial and cytoskeletal function, and were regulated by transcription factors including ARNT2, BHLHE40, FOSL2, JUND, and NFKB1. Conclusions Our results indicate that 25(OH)D during mid-pregnancy, but not at delivery, is associated with placental gene expression at birth. Future research is needed to investigate a potential role of vitamin D in programming placental mitochondrial metabolism, intracellular transport, and transcriptional regulation during pregnancy.
Collapse
Affiliation(s)
- Mariana Parenti
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Melissa M. Melough
- Department of Health Behavior and Nutrition Sciences, University of Delaware, Newark, DE, United States
| | - Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Evan J. Firsick
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Hyo Young Choi
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United States
| | - Karen J. Derefinko
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United States
- Department of Pharmacology, Addiction Science, and Toxicology, University of Tennessee Health Sciences Center, Memphis, TN, United States
| | | | - Kecia N. Carroll
- Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Kaja Z. LeWinn
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
| | - Nicole R. Bush
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Qi Zhao
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, United States
| | - Sheela Sathyanarayana
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
- Department of Epidemiology, University of Washington, Seattle, WA, United States
- Center for Child Health, Behavior, and Development, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| | - Alison G. Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, United States
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
| |
Collapse
|
30
|
Khorami-Sarvestani S, Vanaki N, Shojaeian S, Zarnani K, Stensballe A, Jeddi-Tehrani M, Zarnani AH. Placenta: an old organ with new functions. Front Immunol 2024; 15:1385762. [PMID: 38707901 PMCID: PMC11066266 DOI: 10.3389/fimmu.2024.1385762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
The transition from oviparity to viviparity and the establishment of feto-maternal communications introduced the placenta as the major anatomical site to provide nutrients, gases, and hormones to the developing fetus. The placenta has endocrine functions, orchestrates maternal adaptations to pregnancy at different periods of pregnancy, and acts as a selective barrier to minimize exposure of developing fetus to xenobiotics, pathogens, and parasites. Despite the fact that this ancient organ is central for establishment of a normal pregnancy in eutherians, the placenta remains one of the least studied organs. The first step of pregnancy, embryo implantation, is finely regulated by the trophoectoderm, the precursor of all trophoblast cells. There is a bidirectional communication between placenta and endometrium leading to decidualization, a critical step for maintenance of pregnancy. There are three-direction interactions between the placenta, maternal immune cells, and the endometrium for adaptation of endometrial immune system to the allogeneic fetus. While 65% of all systemically expressed human proteins have been found in the placenta tissues, it expresses numerous placenta-specific proteins, whose expression are dramatically changed in gestational diseases and could serve as biomarkers for early detection of gestational diseases. Surprisingly, placentation and carcinogenesis exhibit numerous shared features in metabolism and cell behavior, proteins and molecular signatures, signaling pathways, and tissue microenvironment, which proposes the concept of "cancer as ectopic trophoblastic cells". By extensive researches in this novel field, a handful of cancer biomarkers has been discovered. This review paper, which has been inspired in part by our extensive experiences during the past couple of years, highlights new aspects of placental functions with emphasis on its immunomodulatory role in establishment of a successful pregnancy and on a potential link between placentation and carcinogenesis.
Collapse
Affiliation(s)
- Sara Khorami-Sarvestani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Negar Vanaki
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorour Shojaeian
- Department of Biochemistry, School of Medical Sciences, Alborz University of Medical Sciences, Karaj, Iran
| | - Kayhan Zarnani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Mahmood Jeddi-Tehrani
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Lai S, Yu W, Liu Y, Yang Y, Zhang X. Current research and evidence gaps on placental development in iron deficiency anemia. Open Life Sci 2024; 19:20220827. [PMID: 38465334 PMCID: PMC10921475 DOI: 10.1515/biol-2022-0827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 03/12/2024] Open
Abstract
Studying the effects of maternal iron deficiency anemia (IDA) is complex owing to its diverse causes, each independently impacting the placenta and fetus. Simple treatment with iron supplements does not always resolve the anemia. Therefore, delving into how IDA alters placental development at a molecular level is crucial to further optimize treatment. This review addresses the effects of IDA on placental structures and functions, including changes in oxygen levels, blood vessels, and the immune system. Profound understanding of physiological characteristics and regulatory mechanisms of placental development is key to explain the mechanisms of abnormal placental development in pregnancy-associated disorders. In turn, future strategies for the prevention and treatment of pregnancy complications involving the placenta can be devised. These studies are significant for improving human reproductive health, enhancing sociodemographic qualities, and even lifelong wellbeing, a focal point in future placental research.
Collapse
Affiliation(s)
- Shaoyang Lai
- Department of Obstetrics, School of Medicine, Women and Children’s Hospital, Xiamen University, Xiamen, China
| | - Weiwei Yu
- Department of Obstetrics, School of Medicine, Women and Children’s Hospital, Xiamen University, Xiamen, China
| | - Ying Liu
- Department of Obstetrics, School of Medicine, Women and Children’s Hospital, Xiamen University, Xiamen, China
| | - Yuxin Yang
- Department of Obstetrics, School of Medicine, Women and Children’s Hospital, Xiamen University, Xiamen, China
| | - Xueqin Zhang
- Department of Obstetrics, School of Medicine, Women and Children’s Hospital, Xiamen University, Xiamen, China
| |
Collapse
|
32
|
Inohaya A, Chigusa Y, Takakura M, Io S, Kim MA, Matsuzaka Y, Yasuda E, Ueda Y, Kawamura Y, Takamatsu S, Mogami H, Takashima Y, Mandai M, Kondoh E. Shear stress in the intervillous space promotes syncytial formation of iPS cells-derived trophoblasts†. Biol Reprod 2024; 110:300-309. [PMID: 37930227 DOI: 10.1093/biolre/ioad143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/01/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023] Open
Abstract
The intervillous space of human placenta is filled with maternal blood, and villous trophoblasts are constantly exposed to the shear stress generated by maternal blood pressure and flow throughout the entire gestation period. However, the effects of shear stress on villous trophoblasts and their biological significance remain unknown. Here, using our recently established naïve human pluripotent stem cells-derived cytotrophoblast stem cells (nCTs) and a device that can apply arbitrary shear stress to cells, we investigated the impact of shear stress on early-stage trophoblasts. After 72 h of exposure to 10 dyn/cm2 shear stress, nCTs became fused and multinuclear, and mRNA expression of the syncytiotrophoblast (ST) markers, such as glial cell missing 1, endogenous retrovirus group W member 1 envelope, chorionic gonadotropin subunit beta 3, syndecan 1, pregnancy specific beta-1-glycoprotein 3, placental growth factor, and solute carrier family 2 member 1 were significantly upregulated compared to static conditions. Immunohistochemistry showed that shear stress increased fusion index, human chorionic gonadotropin secretion, and human placental lactogen secretion. Increased microvilli formation on the surface of nCTs under flow conditions was detected using scanning electron microscopy. Intracellular cyclic adenosine monophosphate significantly increased under flow conditions. Moreover, transcriptome analysis of nCTs subjected to shear stress revealed that shear stress upregulated ST-specific genes and downregulated CT-specific genes. Collectively, these findings indicate that shear stress promotes the differentiation of nCTs into ST.
Collapse
Affiliation(s)
- Asako Inohaya
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshitsugu Chigusa
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahito Takakura
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shingo Io
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Min-A Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yu Matsuzaka
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eriko Yasuda
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Ueda
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yosuke Kawamura
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shiro Takamatsu
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruta Mogami
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuhiro Takashima
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eiji Kondoh
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
33
|
Platt JL, Cascalho M. Somatic Cell Fusion in Host Defense and Adaptation. Results Probl Cell Differ 2024; 71:213-225. [PMID: 37996680 DOI: 10.1007/978-3-031-37936-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Evidence of fusion of somatic cells has been noted in health and in disease for more than a century. The most obvious but uncertain hallmark has been the presence of multiple nuclei in cells. Although multinucleated cells are found in normal and diseased tissues, the benefit or harm of such cells can be difficult to elucidate. Still more difficult however is the identification of mononuclear cells previously formed by fusion of somatic cells with one or more nuclei disposed. The later process can introduce mutations that promote viral diversification, cancer, and tissue senescence. Less obvious the potential benefits of cell fusion. Recent work in cell biology, immunology, and genomic analysis however makes it possible to postulate benefits and potentially arrive at novel therapeutic agents and approaches that replicate or enhance these benefits.
Collapse
Affiliation(s)
- Jeffrey L Platt
- Departments of Surgery and Microbiology & Immunology University of Michigan, Ann Arbor, MI, USA.
| | - Marilia Cascalho
- Departments of Surgery and Microbiology & Immunology University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
34
|
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
|
35
|
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
|
36
|
Khor JM, Guerrero-Santoro J, Ettensohn CA. Molecular compartmentalization in a syncytium: restricted mobility of proteins within the sea urchin skeletogenic mesenchyme. Development 2023; 150:dev201804. [PMID: 37902109 DOI: 10.1242/dev.201804] [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: 03/23/2023] [Accepted: 10/23/2023] [Indexed: 10/31/2023]
Abstract
Multinucleated cells, or syncytia, are found in diverse taxa. Their biological function is often associated with the compartmentalization of biochemical or cellular activities within the syncytium. How such compartments are generated and maintained is poorly understood. The sea urchin embryonic skeleton is secreted by a syncytium, and local patterns of skeletal growth are associated with distinct sub-domains of gene expression within the syncytium. For such molecular compartments to be maintained and to control local patterns of skeletal growth: (1) the mobility of TFs must be restricted to produce stable differences in the transcriptional states of nuclei within the syncytium; and (2) the mobility of biomineralization proteins must also be restricted to produce regional differences in skeletal growth. To test these predictions, we expressed fluorescently tagged forms of transcription factors and biomineralization proteins in sub-domains of the skeletogenic syncytium. We found that both classes of proteins have restricted mobility within the syncytium and identified motifs that limit their mobility. Our findings have general implications for understanding the functional and molecular compartmentalization of syncytia.
Collapse
Affiliation(s)
- Jian Ming Khor
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15218, USA
| | - Jennifer Guerrero-Santoro
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15218, USA
| | - Charles A Ettensohn
- Department of Biological Sciences, Carnegie Mellon University, 4400 Fifth Avenue, Pittsburgh, PA 15218, USA
| |
Collapse
|
37
|
White JS, Su JJ, Ruark EM, Hua J, Hutson MS, Page-McCaw A. Wound-Induced Syncytia Outpace Mononucleate Neighbors during Drosophila Wound Repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.25.546442. [PMID: 37425719 PMCID: PMC10327115 DOI: 10.1101/2023.06.25.546442] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
All organisms have evolved to respond to injury. Cell behaviors like proliferation, migration, and invasion replace missing cells and close wounds. However, the role of other wound-induced cell behaviors is not understood, including the formation of syncytia (multinucleated cells). Wound-induced epithelial syncytia were first reported around puncture wounds in post-mitotic Drosophila epidermal tissues, but have more recently been reported in mitotically competent tissues such as the Drosophila pupal epidermis and zebrafish epicardium. The presence of wound-induced syncytia in mitotically active tissues suggests that syncytia offer adaptive benefits, but it is unknown what those benefits are. Here, we use in vivo live imaging to analyze wound-induced syncytia in mitotically competent Drosophila pupae. We find that almost half the epithelial cells near a wound fuse to form large syncytia. These syncytia use several routes to speed wound repair: they outpace diploid cells to complete wound closure; they reduce cell intercalation during wound closure; and they pool the resources of their component cells to concentrate them toward the wound. In addition to wound healing, these properties of syncytia are likely to contribute to their roles in development and pathology.
Collapse
Affiliation(s)
- James S. White
- Dept. Cell and Developmental Biology, Vanderbilt School of Medicine, Nashville, TN
- Program in Developmental Biology, Vanderbilt University Nashville, TN
| | - Jasmine J. Su
- Dept. Cell and Developmental Biology, Vanderbilt School of Medicine, Nashville, TN
- Dept. Biological Sciences, Vanderbilt University, Nashville, TN
| | - Elizabeth M. Ruark
- Dept. Cell and Developmental Biology, Vanderbilt School of Medicine, Nashville, TN
| | - Junmin Hua
- Dept. Cell and Developmental Biology, Vanderbilt School of Medicine, Nashville, TN
| | - M. Shane Hutson
- Dept. Physics and Astronomy Vanderbilt University Nashville, TN
- Dept. Biological Sciences, Vanderbilt University, Nashville, TN
| | - Andrea Page-McCaw
- Dept. Cell and Developmental Biology, Vanderbilt School of Medicine, Nashville, TN
- Program in Developmental Biology, Vanderbilt University Nashville, TN
- Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN
- Lead Contact
| |
Collapse
|
38
|
García-Montero C, Fraile-Martinez O, De Leon-Oliva D, Boaru DL, Garcia-Puente LM, De León-Luis JA, Bravo C, Diaz-Pedrero R, Lopez-Gonzalez L, Álvarez-Mon M, García-Honduvilla N, Saez MA, Ortega MA. Exploring the Role of Mediterranean and Westernized Diets and Their Main Nutrients in the Modulation of Oxidative Stress in the Placenta: A Narrative Review. Antioxidants (Basel) 2023; 12:1918. [PMID: 38001771 PMCID: PMC10669105 DOI: 10.3390/antiox12111918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Oxidative stress is a major cellular event that occurs in the placenta, fulfilling critical physiological roles in non-pathological pregnancies. However, exacerbated oxidative stress is a pivotal feature of different obstetric complications, like pre-eclampsia, fetal growth restriction, and other diseases. Compelling evidence supports the relevant role of diet during pregnancy, with pleiotropic consequences for maternal well-being. The present review aims to examine the complex background between oxidative stress and placental development and function in physiological conditions, also intending to understand the relationship between different dietary patterns and the human placenta, particularly how this could influence oxidative stress processes. The effects of Westernized diets (WDs) and high-fat diets (HFDs) rich in ultra-processed foods and different additives are compared with healthy patterns such as a Mediterranean diet (MedDiet) abundant in omega 3 polyunsaturated fatty acids, monounsaturated fatty acids, polyphenols, dietary fiber, and vitamins. Although multiple studies have focused on the role of specific nutrients, mostly in animal models and in vitro, further observational and intervention studies focusing on the placental structure and function in women with different dietary patterns should be conducted to understand the precise influence of diet on this organ.
Collapse
Affiliation(s)
- Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Luis M. Garcia-Puente
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Juan A. De León-Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (J.A.D.L.-L.); (C.B.)
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Coral Bravo
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (J.A.D.L.-L.); (C.B.)
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, 28009 Madrid, Spain
- Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Prince of Asturias, Networking Research Center on for Liver and Digestive Diseases (CIBEREHD), 28806 Alcalá de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
- Pathological Anatomy Service, University Hospital Gómez-Ulla, 28806 Alcalá de Henares, Spain
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain; (C.G.-M.); (O.F.-M.); (D.D.L.-O.); (D.L.B.); (L.M.G.-P.); (M.Á.-M.); (N.G.-H.); (M.A.S.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (R.D.-P.); (L.L.-G.)
| |
Collapse
|
39
|
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
|
40
|
de Lima Castro M, Dos Passos RR, Justina VD, do Amaral WN, Giachini FR. Physiological and pathological evidence of O-GlcNAcylation regulation during pregnancy related process. Placenta 2023; 141:43-50. [PMID: 37210277 DOI: 10.1016/j.placenta.2023.04.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/22/2023] [Accepted: 04/25/2023] [Indexed: 05/22/2023]
Abstract
O-GlcNAcylation is a dynamic and reversible post-translational modification (PTM) controlled by the enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Changes in its expression lead to a breakdown in cellular homeostasis, which is linked to several pathological processes. Placentation and embryonic development are periods of high cell activity, and imbalances in cell signaling pathways can result in infertility, miscarriage, or pregnancy complications. O-GlcNAcylation is involved in cellular processes such as genome maintenance, epigenetic regulation, protein synthesis/degradation, metabolic pathways, signaling pathways, apoptosis, and stress response. Trophoblastic differentiation/invasion and placental vasculogenesis, as well as zygote viability and embryonic neuronal development, are all dependent on O-GlcNAcylation. This PTM is required for pluripotency, which is a required condition for embryonic development. Further, this pathway is a nutritional sensor and cell stress marker, which is primarily measured by the OGT enzyme and its product, protein O-GlcNAcylation. Yet, this post-translational modification is enrolled in metabolic and cardiovascular adaptations during pregnancy. Finally, evidence of how O-GlcNAc impacts pregnancy during pathological conditions such as hyperglycemia, gestational diabetes, hypertension, and stress disorders are reviewed. Considering this scenario, progress in understanding the role of O- GlcNAcylation in pregnancy is required.
Collapse
Affiliation(s)
- Marta de Lima Castro
- Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goias, Goiânia, Brazil
| | - Rinaldo Rodrigues Dos Passos
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Vanessa Dela Justina
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Waldemar Naves do Amaral
- Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goias, Goiânia, Brazil
| | - Fernanda Regina Giachini
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil.
| |
Collapse
|
41
|
Yu M, Hu X, Pan Z, Du C, Jiang J, Zheng W, Cai H, Wang Y, Deng W, Wang H, Lu J, Sun MA, Cao B. Endogenous retrovirus-derived enhancers confer the transcriptional regulation of human trophoblast syncytialization. Nucleic Acids Res 2023; 51:4745-4759. [PMID: 36864754 PMCID: PMC10250217 DOI: 10.1093/nar/gkad109] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 03/04/2023] Open
Abstract
Endogenous retroviruses (ERVs) have been proposed as a driving force for the evolution of the mammalian placenta, however, the contribution of ERVs to placental development and the underlying regulatory mechanism remain largely elusive. A key process of placental development is the formation of multinucleated syncytiotrophoblasts (STBs) in direct contact with maternal blood, through which constitutes the maternal-fetal interface critical for nutrient allocation, hormone production and immunological modulation during pregnancy. We delineate that ERVs profoundly rewire the transcriptional program of trophoblast syncytialization. Here, we first determined the dynamic landscape of bivalent ERV-derived enhancers with dual occupancy of H3K27ac and H3K9me3 in human trophoblast stem cells (hTSCs). We further demonstrated that enhancers overlapping several ERV families tend to exhibit increased H3K27ac and reduced H3K9me3 occupancy in STBs relative to hTSCs. Particularly, bivalent enhancers derived from the Simiiformes-specific MER50 transposons were linked to a cluster of genes important for STB formation. Importantly, deletions of MER50 elements adjacent to several STB genes, including MFSD2A and TNFAIP2, significantly attenuated their expression concomitant to compromised syncytium formation. Together, we propose that ERV-derived enhancers, MER50 specifically, fine-tune the transcriptional networks accounting for human trophoblast syncytialization, which sheds light on a novel ERV-mediated regulatory mechanism underlying placental development.
Collapse
Affiliation(s)
- Miao Yu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| | - Xiaoqian Hu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian361002, China
| | - Zihang Pan
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| | - Cui Du
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu225009, China
| | - Jing Jiang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu225009, China
| | - Wanshan Zheng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| | - Han Cai
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| | - Yinan Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| | - Ming-an Sun
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| | - Bin Cao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian361002, China
| |
Collapse
|
42
|
Nashif SK, Mahr RM, Jena S, Jo S, Nelson AB, Sadowski D, Crawford PA, Puchalska P, Alejandro EU, Gearhart MD, Wernimont SA. Metformin impairs trophoblast metabolism and differentiation in a dose-dependent manner. Front Cell Dev Biol 2023; 11:1167097. [PMID: 37250894 PMCID: PMC10213689 DOI: 10.3389/fcell.2023.1167097] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/05/2023] [Indexed: 05/31/2023] Open
Abstract
Metformin is a widely prescribed medication whose mechanism of action is not completely defined and whose role in gestational diabetes management remains controversial. In addition to increasing the risk of fetal growth abnormalities and preeclampsia, gestational diabetes is associated with abnormalities in placental development including impairments in trophoblast differentiation. Given that metformin impacts cellular differentiation events in other systems, we assessed metformin's impact on trophoblast metabolism and differentiation. Using established cell culture models of trophoblast differentiation, oxygen consumption rates and relative metabolite abundance were determined following 200 µM (therapeutic range) and 2000 µM (supra-therapeutic range) metformin treatment using Seahorse and mass-spectrometry approaches. While no differences in oxygen consumption rates or relative metabolite abundance were detected between vehicle and 200 µM metformin-treated cells, 2000 µM metformin impaired oxidative metabolism and increased the abundance of lactate and TCA cycle intermediates, α-ketoglutarate, succinate, and malate. Examining differentiation, treatment with 2000 μM, but not 200 µM metformin, impaired HCG production and expression of multiple trophoblast differentiation markers. Overall, this work suggests that supra-therapeutic concentrations of metformin impair trophoblast metabolism and differentiation whereas metformin concentrations in the therapeutic range do not strongly impact these processes.
Collapse
Affiliation(s)
- Sereen K. Nashif
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, United States
| | - Renee M. Mahr
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, United States
| | - Snehalata Jena
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, United States
| | - Seokwon Jo
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Alisa B. Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Danielle Sadowski
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, United States
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Emilyn U. Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Micah D. Gearhart
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, United States
| | - Sarah A. Wernimont
- Department of Obstetrics, Gynecology and Women’s Health, University of Minnesota, Minneapolis, MN, United States
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
43
|
Mahr RM, Jena S, Nashif SK, Nelson AB, Rauckhorst AJ, Rome FI, Sheldon RD, Hughey CC, Puchalska P, Gearhart MD, Taylor EB, Crawford PA, Wernimont SA. Mitochondrial citrate metabolism and efflux regulate BeWo differentiation. Sci Rep 2023; 13:7387. [PMID: 37149697 PMCID: PMC10164164 DOI: 10.1038/s41598-023-34435-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 04/29/2023] [Indexed: 05/08/2023] Open
Abstract
Cytotrophoblasts fuse to form and renew syncytiotrophoblasts necessary to maintain placental health throughout gestation. During cytotrophoblast to syncytiotrophoblast differentiation, cells undergo regulated metabolic and transcriptional reprogramming. Mitochondria play a critical role in differentiation events in cellular systems, thus we hypothesized that mitochondrial metabolism played a central role in trophoblast differentiation. In this work, we employed static and stable isotope tracing untargeted metabolomics methods along with gene expression and histone acetylation studies in an established BeWo cell culture model of trophoblast differentiation. Differentiation was associated with increased abundance of the TCA cycle intermediates citrate and α-ketoglutarate. Citrate was preferentially exported from mitochondria in the undifferentiated state but was retained to a larger extent within mitochondria upon differentiation. Correspondingly, differentiation was associated with decreased expression of the mitochondrial citrate transporter (CIC). CRISPR/Cas9 disruption of the mitochondrial citrate carrier showed that CIC is required for biochemical differentiation of trophoblasts. Loss of CIC resulted in broad alterations in gene expression and histone acetylation. These gene expression changes were partially rescued through acetate supplementation. Taken together, these results highlight a central role for mitochondrial citrate metabolism in orchestrating histone acetylation and gene expression during trophoblast differentiation.
Collapse
Affiliation(s)
- Renee M Mahr
- Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Snehalata Jena
- Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Sereen K Nashif
- Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Alisa B Nelson
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Adam J Rauckhorst
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Ferrol I Rome
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Ryan D Sheldon
- Department of Biochemistry, University of Iowa, Iowa City, IA, USA
| | - Curtis C Hughey
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Patrycja Puchalska
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Micah D Gearhart
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Eric B Taylor
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| | - Peter A Crawford
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Sarah A Wernimont
- Department of Obstetrics, Gynecology and Women's Health, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA.
- Division of Molecular Medicine, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
44
|
Trinh QD, Pham NTK, Takada K, Ushijima H, Komine-Aizawa S, Hayakawa S. Roles of TGF-β1 in Viral Infection during Pregnancy: Research Update and Perspectives. Int J Mol Sci 2023; 24:ijms24076489. [PMID: 37047462 PMCID: PMC10095195 DOI: 10.3390/ijms24076489] [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: 03/04/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
Transforming growth factor-beta 1 (TGF-β1) is a pleiotropic growth factor playing various roles in the human body including cell growth and development. More functions of TGF-β1 have been discovered, especially its roles in viral infection. TGF-β1 is abundant at the maternal-fetal interface during pregnancy and plays an important function in immune tolerance, an essential key factor for pregnancy success. It plays some critical roles in viral infection in pregnancy, such as its effects on the infection and replication of human cytomegalovirus in syncytiotrophoblasts. Interestingly, its role in the enhancement of Zika virus (ZIKV) infection and replication in first-trimester trophoblasts has recently been reported. The above up-to-date findings have opened one of the promising approaches to studying the mechanisms of viral infection during pregnancy with links to corresponding congenital syndromes. In this article, we review our current and recent advances in understanding the roles of TGF-β1 in viral infection. Our discussion focuses on viral infection during pregnancy, especially in the first trimester. We highlight the mutual roles of viral infection and TGF-β1 in specific contexts and possible functions of the Smad pathway in viral infection, with a special note on ZIKV infection. In addition, we discuss promising approaches to performing further studies on this topic.
Collapse
Affiliation(s)
- Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
45
|
Nashif SK, Mahr RM, Jena S, Jo S, Nelson AB, Sadowski D, Crawford PA, Puchalska P, Alejandro EU, Gearhart MD, Wernimont SA. Metformin impairs trophoblast metabolism and differentiation in dose dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.14.528531. [PMID: 36824783 PMCID: PMC9949099 DOI: 10.1101/2023.02.14.528531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Metformin is a widely prescribed medication whose mechanism of action is not completely defined and whose role in gestational diabetes management remains controversial. In addition to increasing risks of fetal growth abnormalities and preeclampsia, gestational diabetes is associated with abnormalities in placental development including impairments in trophoblast differentiation. Given that metformin impacts cellular differentiation events in other systems, we assessed metformin's impact on trophoblast metabolism and differentiation. Using established cell culture models of trophoblast differentiation, oxygen consumption rates and relative metabolite abundance were determined following 200 μM (therapeutic range) and 2000 μM (supra-therapeutic range) metformin treatment using Seahorse and mass-spectrometry approaches. While no differences in oxygen consumption rates or relative metabolite abundance were detected between vehicle and 200 μM metformin treated cells, 2000 μM metformin impaired oxidative metabolism and increased abundance of lactate and TCA cycle intermediates, α-ketoglutarate, succinate, and malate. Examining differentiation, treatment with 2000 μM, but not 200 μM metformin, impaired HCG production and expression of multiple trophoblast differentiation markers. Overall, this work suggests that supra-therapeutic concentrations of metformin impairs trophoblast metabolism and differentiation whereas metformin concentrations in the therapeutic range do not strongly impact these processes.
Collapse
|
46
|
Mahr RM, Jena S, Nashif SK, Nelson AB, Rauckhorst AJ, Rome FI, Sheldon RD, Hughey CC, Puchalska P, Gearhart MD, Taylor EB, Crawford PA, Wernimont SA. Mitochondrial citrate metabolism and efflux regulates trophoblast differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525071. [PMID: 36711862 PMCID: PMC9882289 DOI: 10.1101/2023.01.22.525071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Cytotrophoblasts fuse to form and renew syncytiotrophoblasts necessary to maintain placental health throughout gestation. During cytotrophoblast to syncytiotrophoblast differentiation, cells undergo regulated metabolic and transcriptional reprogramming. Mitochondria play a critical role in differentiation events in cellular systems, thus we hypothesized that mitochondrial metabolism played a central role in trophoblast differentiation. In this work, we employed static and stable isotope tracing untargeted metabolomics methods along with gene expression and histone acetylation studies in an established cell culture model of trophoblast differentiation. Trophoblast differentiation was associated with increased abundance of the TCA cycle intermediates citrate and α-ketoglutarate. Citrate was preferentially exported from mitochondria in the undifferentiated state but was retained to a larger extent within mitochondria upon differentiation. Correspondingly, differentiation was associated with decreased expression of the mitochondrial citrate transporter (CIC). CRISPR/Cas9 disruption of the mitochondrial citrate carrier showed that CIC is required for biochemical differentiation of trophoblasts. Loss of CIC resulted in broad alterations in gene expression and histone acetylation. These gene expression changes were partially rescued through acetate supplementation. Taken together, these results highlight a central role for mitochondrial citrate metabolism in orchestrating histone acetylation and gene expression during trophoblast differentiation.
Collapse
|
47
|
Chenge S, Ngure H, Kanoi BN, Sferruzzi-Perri AN, Kobia FM. Infectious and environmental placental insults: from underlying biological pathways to diagnostics and treatments. Pathog Dis 2023; 81:ftad024. [PMID: 37727973 DOI: 10.1093/femspd/ftad024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/15/2023] [Accepted: 09/18/2023] [Indexed: 09/21/2023] Open
Abstract
Because the placenta is bathed in maternal blood, it is exposed to infectious agents and chemicals that may be present in the mother's circulation. Such exposures, which do not necessarily equate with transmission to the fetus, may primarily cause placental injury, thereby impairing placental function. Recent research has improved our understanding of the mechanisms by which some infectious agents are transmitted to the fetus, as well as the mechanisms underlying their impact on fetal outcomes. However, less is known about the impact of placental infection on placental structure and function, or the mechanisms underlying infection-driven placental pathogenesis. Moreover, recent studies indicate that noninfectious environmental agents accumulate in the placenta, but their impacts on placental function and fetal outcomes are unknown. Critically, diagnosing placental insults during pregnancy is very difficult and currently, this is possible only through postpartum placental examination. Here, with emphasis on humans, we discuss what is known about the impact of infectious and chemical agents on placental physiology and function, particularly in the absence of maternal-fetal transmission, and highlight knowledge gaps with potential implications for diagnosis and intervention against placental pathologies.
Collapse
Affiliation(s)
- Samuel Chenge
- Department of Medical Microbiology and Laboratory Sciences, Jomo Kenyatta University of Agriculture and Technology, Juja, off Thika road, P. O. Box 62000-00200 Nairobi, Kenya
| | - Harrison Ngure
- Directorate of Research and Innovation, Mount Kenya University, General Kago road, P.O. Box 342-01000, Thika, Kenya
| | - Bernard N Kanoi
- Directorate of Research and Innovation, Mount Kenya University, General Kago road, P.O. Box 342-01000, Thika, Kenya
- Centre for Malaria Elimination, Mount Kenya University, General Kago road, P.O. Box 342-01000, Thika, Kenya
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Francis M Kobia
- Directorate of Research and Innovation, Mount Kenya University, General Kago road, P.O. Box 342-01000, Thika, Kenya
- Centre for Malaria Elimination, Mount Kenya University, General Kago road, P.O. Box 342-01000, Thika, Kenya
| |
Collapse
|