1
|
Sturgill I, Raab J, Hoadley K. Expanded detection and impact of BAP1 alterations in cancer. NAR Cancer 2024; 6:zcae045. [PMID: 39554490 PMCID: PMC11567159 DOI: 10.1093/narcan/zcae045] [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: 07/17/2024] [Revised: 09/15/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024] Open
Abstract
Aberrant expression of the BAP1 (BRCA associated protein 1) tumor suppressor gene is a prominent risk factor for several tumor types and is important in tumor evolution and progression. Here we performed integrated multi-omics analyses using data from The Cancer Genome Atlas for 33 cancer types and over 10 000 individuals to identify alterations leading to BAP1 disruption. We combined existing variant calls and new calls derived from a de novo local realignment pipeline across multiple independent variant callers, increasing somatic variant detection by 41% from 182 to 257, including 11 indels ≥40 bp. The expanded detection of mutations highlights the power of new tools to uncover longer indels and impactful mutations. We developed an expression-based BAP1 activity score and identified a transcriptional profile associated with BAP1 disruption in cancer. BAP1 has been proposed to play a critical role in controlling tumor plasticity and normal cell fate. Leveraging human and mouse liver datasets, BAP1 loss in normal cells resulted in lower BAP1 activity scores and lower scores were associated with a less-differentiated phenotype in embryonic cells. Together, our expanded BAP1 mutant samples revealed a transcriptional signature in cancer cells, supporting BAP1's influences on cellular plasticity and cell identity maintenance.
Collapse
Affiliation(s)
- Ian R Sturgill
- Bioinformatics and Computational Biology Curriculum, Department of Genetics, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| | - Jesse R Raab
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| | - Katherine A Hoadley
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC 27599, USA
| |
Collapse
|
2
|
Lizárraga-Verdugo E, Beltrán-Ontiveros SA, Gutiérrez-Grijalva EP, Montoya-Moreno M, Gutiérrez-Arzapalo PY, Avendaño-Félix M, Gutiérrez-Castro KP, Cuén-Lazcano DE, González-Quintero P, Mora-Palazuelos CE. The Underlying Molecular Mechanisms of the Placenta Accreta Spectrum: A Narrative Review. Int J Mol Sci 2024; 25:9722. [PMID: 39273667 PMCID: PMC11395310 DOI: 10.3390/ijms25179722] [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: 07/12/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Placenta accreta spectrum (PAS) disorders are characterized by abnormal trophoblastic invasion into the myometrium, leading to significant maternal health risks. PAS includes placenta accreta (invasion < 50% of the myometrium), increta (invasion > 50%), and percreta (invasion through the entire myometrium). The condition is most associated with previous cesarean deliveries and increases in chance with the number of prior cesarians. The increasing global cesarean rates heighten the importance of early PAS diagnosis and management. This review explores genetic expression and key regulatory processes, such as apoptosis, cell proliferation, invasion, and inflammation, focusing on signaling pathways, genetic expression, biomarkers, and non-coding RNAs involved in trophoblastic invasion. It compiles the recent scientific literature (2014-2024) from the Scopus, PubMed, Google Scholar, and Web of Science databases. Identifying new biomarkers like AFP, sFlt-1, β-hCG, PlGF, and PAPP-A aids in early detection and management. Understanding genetic expression and non-coding RNAs is crucial for unraveling PAS complexities. In addition, aberrant signaling pathways like Notch, PI3K/Akt, STAT3, and TGF-β offer potential therapeutic targets to modulate trophoblastic invasion. This review underscores the need for interdisciplinary care, early diagnosis, and ongoing research into PAS biomarkers and molecular mechanisms to improve prognosis and quality of life for affected women.
Collapse
Affiliation(s)
- Erik Lizárraga-Verdugo
- Research Unit, Center for Research and Teaching in Health Sciences, Autonomous University of Sinaloa, Culiacan 80030, Mexico
| | - Saúl Armando Beltrán-Ontiveros
- Research Unit, Center for Research and Teaching in Health Sciences, Autonomous University of Sinaloa, Culiacan 80030, Mexico
| | | | - Marisol Montoya-Moreno
- Research Unit, Center for Research and Teaching in Health Sciences, Autonomous University of Sinaloa, Culiacan 80030, Mexico
| | - Perla Y Gutiérrez-Arzapalo
- Research Unit, Center for Research and Teaching in Health Sciences, Autonomous University of Sinaloa, Culiacan 80030, Mexico
| | | | - Karla Paola Gutiérrez-Castro
- Research Unit, Center for Research and Teaching in Health Sciences, Autonomous University of Sinaloa, Culiacan 80030, Mexico
| | - Daniel E Cuén-Lazcano
- Research Unit, Center for Research and Teaching in Health Sciences, Autonomous University of Sinaloa, Culiacan 80030, Mexico
| | - Paul González-Quintero
- Gynecology and Obstetrics Service, Women's Hospital of Culiacan, Health Secretary, Culiacan 80020, Mexico
| | - Carlos Ernesto Mora-Palazuelos
- Research Unit, Center for Research and Teaching in Health Sciences, Autonomous University of Sinaloa, Culiacan 80030, Mexico
| |
Collapse
|
3
|
Doria-Borrell P, Pérez-García V. Understanding the intersection between placental development and cancer: Lessons from the tumor suppressor BAP1. Commun Biol 2024; 7:1053. [PMID: 39191942 PMCID: PMC11349880 DOI: 10.1038/s42003-024-06689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
The placenta, a pivotal organ in mammalian reproduction, allows nutrient exchange and hormonal signaling between the mother and the developing fetus. Understanding its molecular intricacies is essential for deciphering normal embryonic development and pathological conditions such as tumorigenesis. Here, we explore the multifaceted role of the tumor suppressor BRCA1-associated protein 1 (BAP1) in cancer and placentation. Initially recognized for its tumor-suppressive properties, BAP1 has emerged as a key regulator at the intersection of tumorigenesis and placental development. BAP1 influences crucial cellular processes such as cell death, proliferation, metabolism, and response to hypoxic conditions. By integrating insights from tumor and developmental biology, we illuminate the complex molecular pathways orchestrated by BAP1. This perspective highlights BAP1's significant impact on both cancer and placental development, and suggests novel therapeutic strategies that could improve outcomes for pregnancy disorders and cancer.
Collapse
Affiliation(s)
| | - Vicente Pérez-García
- Centro de Investigación Príncipe Felipe, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| |
Collapse
|
4
|
Walewska E, Makowczenko KG, Witek K, Laniecka E, Molcan T, Alvarez-Sanchez A, Kelsey G, Perez-Garcia V, Galvão AM. Fetal growth restriction and placental defects in obese mice are associated with impaired decidualisation: the role of increased leptin signalling modulators SOCS3 and PTPN2. Cell Mol Life Sci 2024; 81:329. [PMID: 39090270 PMCID: PMC11335253 DOI: 10.1007/s00018-024-05336-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 04/05/2024] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Abstract
Decidualisation of the endometrium is a key event in early pregnancy, which enables embryo implantation. Importantly, the molecular processes impairing decidualisation in obese mothers are yet to be characterised. We hypothesise that impaired decidualisation in obese mice is mediated by the upregulation of leptin modulators, the suppressor of cytokine signalling 3 (SOCS3) and the protein tyrosine phosphatase non-receptor type 2 (PTPN2), together with the disruption of progesterone (P4)-signal transducer and activator of transcription (STAT3) signalling. After feeding mice with chow diet (CD) or high-fat diet (HFD) for 16 weeks, we confirmed the downregulation of P4 and oestradiol (E2) steroid receptors in decidua from embryonic day (E) 6.5 and decreased proliferation of stromal cells from HFD. In vitro decidualised mouse endometrial stromal cells (MESCs) and E6.5 deciduas from the HFD showed decreased expression of decidualisation markers, followed by the upregulation of SOCS3 and PTPN2 and decreased phosphorylation of STAT3. In vivo and in vitro leptin treatment of mice and MESCs mimicked the results observed in the obese model. The downregulation of Socs3 and Ptpn2 after siRNA transfection of MESCs from HFD mice restored the expression level of decidualisation markers. Finally, DIO mice placentas from E18.5 showed decreased labyrinth development and vascularisation and fetal growth restricted embryos. The present study revealed major defects in decidualisation in obese mice, characterised by altered uterine response to E2 and P4 steroid signalling. Importantly, altered hormonal response was associated with increased expression of leptin signalling modulators SOCS3 and PTPN2. Elevated levels of SOCS3 and PTPN2 were shown to molecularly affect decidualisation in obese mice, potentially disrupting the STAT3-PR regulatory molecular hub.
Collapse
Affiliation(s)
- Edyta Walewska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Karol G Makowczenko
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Krzysztof Witek
- Laboratory of Cell and Tissue Analysis and Imaging, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Elżbieta Laniecka
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Tomasz Molcan
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland
| | - Andrea Alvarez-Sanchez
- Molecular Mechanisms of Placental Invasion, Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, 46012, Valencia, Spain
| | - Gavin Kelsey
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
| | - Vicente Perez-Garcia
- Molecular Mechanisms of Placental Invasion, Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera 3, 46012, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), 28049, Madrid, Spain.
| | - António M Galvão
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Olsztyn, Poland.
- Epigenetics Programme, The Babraham Institute, Cambridge, CB22 3AT, UK.
- Centre for Trophoblast Research, University of Cambridge, Cambridge, CB2 3EG, UK.
- Department of Comparative Biomedical Sciences, Royal Veterinary College, 4 Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
5
|
Varberg KM, Moreno-Irusta A, Novoa A, Musser B, Varberg JM, Goering JP, Saadi I, Iqbal K, Okae H, Arima T, Williams J, Pisarska MD, Soares MJ. Leveraging chorionic villus biopsies for the derivation of patient-specific trophoblast stem cells. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2022.12.07.22283218. [PMID: 39108523 PMCID: PMC11302605 DOI: 10.1101/2022.12.07.22283218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Human trophoblast stem (TS) cells are an informative in vitro model for the generation and testing of biologically meaningful hypotheses. The goal of this project was to derive patient-specific TS cell lines from clinically available chorionic villus sampling biopsies. Cell outgrowths were captured from human chorionic villus tissue specimens cultured in modified human TS cell medium. Cell colonies emerged early during the culture and cell lines were established and passaged for several generations. Karyotypes of the newly established chorionic villus-derived trophoblast stem (TS CV ) cell lines were determined and compared to initial genetic diagnoses from freshly isolated chorionic villi. Phenotypes of TSCV cells in the stem state and following differentiation were compared to cytotrophoblast-derived TS (TS CT ) cells. TSCV and TSCT cells uniformly exhibited similarities in the stem state and following differentiation into syncytiotrophoblast and extravillous trophoblast cells. Chorionic villus tissue specimens provide a valuable source for TS cell derivation. They expand the genetic diversity of available TS cells and are associated with defined clinical outcomes. TSCV cell lines provide a new set of experimental tools for investigating trophoblast cell lineage development.
Collapse
Affiliation(s)
- Kaela M. Varberg
- 1nstitute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Ayelen Moreno-Irusta
- 1nstitute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Allynson Novoa
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Brynne Musser
- 1nstitute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | | | - Jeremy P. Goering
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Irfan Saadi
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160
| | - Khursheed Iqbal
- 1nstitute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
| | - Hiroaki Okae
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - John Williams
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA
- David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Margareta D. Pisarska
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA
- David Geffen School of Medicine, University of California, Los Angeles, CA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Michael J. Soares
- 1nstitute for Reproductive and Developmental Sciences, Department of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, KS 66160
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160
| |
Collapse
|
6
|
Sturgill IR, Raab JR, Hoadley KA. Expanded detection and impact of BAP1 alterations in cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.21.568094. [PMID: 38045292 PMCID: PMC10690206 DOI: 10.1101/2023.11.21.568094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Aberrant expression of the BAP1 tumor suppressor gene is a prominent risk factor for several tumor types and is important in tumor evolution and progression. Here we performed integrated multi-omic analyses using data from The Cancer Genome Atlas (TCGA) for 33 cancer types and over 10,000 individuals to identify alterations leading to BAP1 disruption. We combined existing variant calls and new calls derived from a de novo local realignment pipeline across multiple independent variant callers, increasing somatic variant detection by 41% from 182 to 257, including 11 indels ≥40bp. The expanded detection of mutations highlights the power of new tools to uncover longer indels and impactful mutations. We developed an expression-based BAP1 activity score and identified a transcriptional profile associated with BAP1 disruption in cancer. BAP1 has been proposed to play a critical role in controlling tumor plasticity and normal cell fate. Leveraging human and mouse liver datasets, BAP1 loss in normal cells resulted in lower BAP1 activity scores and lower scores were associated with a less-differentiated phenotype in embryonic cells. Together, our expanded BAP1 mutant samples revealed a transcriptional signature in cancer cells, supporting BAP1's influences on cellular plasticity and cell identity maintenance.
Collapse
|
7
|
Shastry S, Samal D, Pethe P. Histone H2A deubiquitinase BAP1 is essential for endothelial cell differentiation from human pluripotent stem cells. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-00935-x. [PMID: 38976206 DOI: 10.1007/s11626-024-00935-x] [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: 02/13/2024] [Accepted: 05/25/2024] [Indexed: 07/09/2024]
Abstract
Polycomb group proteins (PcGs) add repressive post translational histone modifications such as H2AK119ub1, and histone H2A deubiquitinases remove it. Mice lacking histone H2A deubiquitinases such as Usp16 and Bap1 die in embryonic stage, while mice lacking Usp3, Mysm1, Usp12, and Usp21 have been shown to be deficient in hematopoietic lineage differentiation, cell cycle regulation, and DNA repair. Thus, it is likely that histone deubiquitinases may also be required for human endothelial cell differentiation; however, there are no reports about the role of histone H2A deubiquitinase BAP1 in human endothelial cell development. We differentiated human pluripotent stem cells into the endothelial lineage which expressed stable inducible shRNA against BAP1. Our results show that BAP1 is required for human endothelial cell differentiation.
Collapse
Affiliation(s)
- Shruti Shastry
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
- Worcester Polytechnic Institute (WPI), Boston, USA
| | - Dharitree Samal
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India
| | - Prasad Pethe
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International (Deemed University), Pune, India.
| |
Collapse
|
8
|
Sonnemann HM, Pazdrak B, Nassif B, Sun Y, Elzohary L, Talukder AH, Katailiha AS, Bhat K, Lizée G. Placental co-transcriptional activator Vestigial-like 1 (VGLL1) drives tumorigenesis via increasing transcription of proliferation and invasion genes. Front Oncol 2024; 14:1403052. [PMID: 38912065 PMCID: PMC11190739 DOI: 10.3389/fonc.2024.1403052] [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: 03/18/2024] [Accepted: 05/10/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction Vestigial-like 1 (VGLL1) is a co-transcriptional activator that binds to TEA domain-containing transcription factors (TEADs). Its expression is upregulated in a variety of aggressive cancer types, including pancreatic and basal-like breast cancer, and increased transcription of VGLL1 is strongly correlated with poor prognosis and decreased overall patient survival. In normal tissues, VGLL1 is most highly expressed within placental trophoblast cells, which share the common attributes of rapid cellular proliferation and invasion with tumor cells. The impact of VGLL1 in cancer has not been fully elucidated and no VGLL1-targeted therapy currently exists. Methods The aim of this study was to evaluate the cellular function and downstream genomic targets of VGLL1 in placental, pancreatic, and breast cancer cells. Functional assays were employed to assess the role of VGLL1 in cellular invasion and proliferation, and ChIP-seq and RNAseq assays were performed to identify VGLL1 target genes and potential impact using pathway analysis. Results ChIP-seq analysis identified eight transcription factors with a VGLL1-binding motif that were common between all three cell types, including TEAD1-4, AP-1, and GATA6, and revealed ~3,000 shared genes with which VGLL1 interacts. Furthermore, increased VGLL1 expression led to an enhancement of cell invasion and proliferation, which was supported by RNAseq analysis showing transcriptional changes in several genes known to be involved in these processes. Discussion This work expands our mechanistic understanding of VGLL1 function in tumor cells and provides a strong rationale for developing VGLL1-targeted therapies for treating cancer patients.
Collapse
Affiliation(s)
- Heather M. Sonnemann
- University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
- Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, United States
| | - Barbara Pazdrak
- Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, United States
| | - Barbara Nassif
- University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
- Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, United States
| | - Yimo Sun
- University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX, United States
- Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, United States
| | - Lama Elzohary
- Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, United States
| | - Amjad H. Talukder
- Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, United States
| | - Arjun S. Katailiha
- Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, United States
| | - Krishna Bhat
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center, Houston, TX, United States
| | - Gregory Lizée
- Department of Melanoma Medical Oncology, UT MD Anderson Cancer Center, Houston, TX, United States
- Department of Immunology, UT MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
9
|
Álvarez-Sánchez A, Grinat J, Doria-Borrell P, Mellado-López M, Pedrera-Alcócer É, Malenchini M, Meseguer S, Hemberger M, Pérez-García V. The GPI-anchor biosynthesis pathway is critical for syncytiotrophoblast differentiation and placental development. Cell Mol Life Sci 2024; 81:246. [PMID: 38819479 PMCID: PMC11143174 DOI: 10.1007/s00018-024-05284-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024]
Abstract
The glycosylphosphatidylinositol (GPI) biosynthetic pathway in the endoplasmic reticulum (ER) is crucial for generating GPI-anchored proteins (GPI-APs), which are translocated to the cell surface and play a vital role in cell signaling and adhesion. This study focuses on two integral components of the GPI pathway, the PIGL and PIGF proteins, and their significance in trophoblast biology. We show that GPI pathway mutations impact on placental development impairing the differentiation of the syncytiotrophoblast (SynT), and especially the SynT-II layer, which is essential for the establishment of the definitive nutrient exchange area within the placental labyrinth. CRISPR/Cas9 knockout of Pigl and Pigf in mouse trophoblast stem cells (mTSCs) confirms the role of these GPI enzymes in syncytiotrophoblast differentiation. Mechanistically, impaired GPI-AP generation induces an excessive unfolded protein response (UPR) in the ER in mTSCs growing in stem cell conditions, akin to what is observed in human preeclampsia. Upon differentiation, the impairment of the GPI pathway hinders the induction of WNT signaling for early SynT-II development. Remarkably, the transcriptomic profile of Pigl- and Pigf-deficient cells separates human patient placental samples into preeclampsia and control groups, suggesting an involvement of Pigl and Pigf in establishing a preeclamptic gene signature. Our study unveils the pivotal role of GPI biosynthesis in early placentation and uncovers a new preeclampsia gene expression profile associated with mutations in the GPI biosynthesis pathway, providing novel molecular insights into placental development with implications for enhanced patient stratification and timely interventions.
Collapse
Affiliation(s)
- Andrea Álvarez-Sánchez
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Johanna Grinat
- Epigenetics Programme, The Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Paula Doria-Borrell
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Maravillas Mellado-López
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Érica Pedrera-Alcócer
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Marta Malenchini
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Salvador Meseguer
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain
| | - Myriam Hemberger
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Canada
| | - Vicente Pérez-García
- Centro de Investigación Príncipe Felipe, Calle de Eduardo Primo Yúfera, 3, 46012, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| |
Collapse
|
10
|
Kim S, Lee EW, Oh DB, Seo J. BAP1 controls mesenchymal stem cell migration by inhibiting the ERK signaling pathway. BMB Rep 2024; 57:250-255. [PMID: 37964637 PMCID: PMC11139679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/19/2023] [Accepted: 11/09/2023] [Indexed: 11/16/2023] Open
Abstract
Due to their stem-like characteristics and immunosuppressive properties, Mesenchymal stem cells (MSCs) offer remarkable potential in regenerative medicine. Much effort has been devoted to enhancing the efficacy of MSC therapy by enhancing MSC migration. In this study, we identified deubiquitinase BRCA1- associated protein 1 (BAP1) as an inhibitor of MSC migration. Using deubiquitinase siRNA library screening based on an in vitro wound healing assay, we found that silencing BAP1 significantly augmented MSC migration. Conversely, BAP1 overexpression reduced the migration and invasion capabilities of MSCs. BAP1 depletion in MSCs upregulates ERK phosphorylation, thereby increasing the expression of the migration factor, osteopontin. Further examination revealed that BAP1 interacts with phosphorylated ERK1/2, deubiquitinating their ubiquitins, and thus attenuating the ERK signaling pathway. Overall, our study highlights the critical role of BAP1 in regulating MSC migration through its deubiquitinase activity, and suggests a novel approach to improve the therapeutic potential of MSCs in regenerative medicine. [BMB Reports 2024; 57(5): 250-255].
Collapse
Affiliation(s)
- Seobin Kim
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Eun-Woo Lee
- Metabolic Disease Research Center, KRIBB, Daejeon 34141, Korea
- Department of Functional Genomics, UST, Daejeon 34113, Korea
| | - Doo-Byoung Oh
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Jinho Seo
- Aging Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biosystems and Bioengineering, University of Science and Technology (UST), Daejeon 34113, Korea
| |
Collapse
|
11
|
Azagury M, Buganim Y. Unlocking trophectoderm mysteries: In vivo and in vitro perspectives on human and mouse trophoblast fate induction. Dev Cell 2024; 59:941-960. [PMID: 38653193 DOI: 10.1016/j.devcel.2024.03.029] [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: 09/07/2023] [Revised: 11/10/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024]
Abstract
In recent years, the pursuit of inducing the trophoblast stem cell (TSC) state has gained prominence as a compelling research objective, illuminating the establishment of the trophoblast lineage and unlocking insights into early embryogenesis. In this review, we examine how advancements in diverse technologies, including in vivo time course transcriptomics, cellular reprogramming to TSC state, chemical induction of totipotent stem-cell-like state, and stem-cell-based embryo-like structures, have enriched our insights into the intricate molecular mechanisms and signaling pathways that define the mouse and human trophectoderm/TSC states. We delve into disparities between mouse and human trophectoderm/TSC fate establishment, with a special emphasis on the intriguing role of pluripotency in this context. Additionally, we re-evaluate recent findings concerning the potential of totipotent-stem-like cells and embryo-like structures to fully manifest the trophectoderm/trophoblast lineage's capabilities. Lastly, we briefly discuss the potential applications of induced TSCs in pregnancy-related disease modeling.
Collapse
Affiliation(s)
- Meir Azagury
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Yosef Buganim
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel.
| |
Collapse
|
12
|
Ryan CW, Peirent ER, Regan SL, Guxholli A, Bielas SL. H2A monoubiquitination: insights from human genetics and animal models. Hum Genet 2024; 143:511-527. [PMID: 37086328 DOI: 10.1007/s00439-023-02557-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/10/2023] [Indexed: 04/23/2023]
Abstract
Metazoan development arises from spatiotemporal control of gene expression, which depends on epigenetic regulators like the polycomb group proteins (PcG) that govern the chromatin landscape. PcG proteins facilitate the addition and removal of histone 2A monoubiquitination at lysine 119 (H2AK119ub1), which regulates gene expression, cell fate decisions, cell cycle progression, and DNA damage repair. Regulation of these processes by PcG proteins is necessary for proper development, as pathogenic variants in these genes are increasingly recognized to underly developmental disorders. Overlapping features of developmental syndromes associated with pathogenic variants in specific PcG genes suggest disruption of central developmental mechanisms; however, unique clinical features observed in each syndrome suggest additional non-redundant functions for each PcG gene. In this review, we describe the clinical manifestations of pathogenic PcG gene variants, review what is known about the molecular functions of these gene products during development, and interpret the clinical data to summarize the current evidence toward an understanding of the genetic and molecular mechanism.
Collapse
Affiliation(s)
- Charles W Ryan
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109-5618, USA
- Medical Science Training Program, University of Michigan Medical School, 3703 Med Sci II, 1241 E. Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Emily R Peirent
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109-5618, USA
| | - Samantha L Regan
- Department of Human Genetics, University of Michigan Medical School, 3703 Med Sci II, 1241 E. Catherine St., Ann Arbor, MI, 48109-5618, USA
| | - Alba Guxholli
- Department of Human Genetics, University of Michigan Medical School, 3703 Med Sci II, 1241 E. Catherine St., Ann Arbor, MI, 48109-5618, USA
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, 48199-5618, USA
| | - Stephanie L Bielas
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109-5618, USA.
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, 48109-5618, USA.
- Department of Human Genetics, University of Michigan Medical School, 3703 Med Sci II, 1241 E. Catherine St., Ann Arbor, MI, 48109-5618, USA.
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, 48199-5618, USA.
| |
Collapse
|
13
|
Chen H, Li R, Bian J, Li X, Su C, Wang Y, Zhang H, Zheng J, Wang Y, Zhang H. OLFML3 suppresses trophoblast apoptosis via the PI3K/AKT pathway: A possible therapeutic target in preeclampsia. Placenta 2024; 147:1-11. [PMID: 38277999 DOI: 10.1016/j.placenta.2024.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
INTRODUCTION Preeclampsia (PE) is a pregnancy complication that encompasses various pathogenic mechanisms. Shallow implantation of the placenta due to abnormal trophoblast behavior is considered an important mechanism underlying PE; however, its exact etiology remains unclear. METHODS The expression of OLFML3 in the placenta and important clinical indicators were performed, followed by a correlation analysis. The effect of OLFML3 on the behavior of HTR-8/SVneo cells was examined, and the downstream molecular mechanisms of OLFML3 were investigated in HTR-8/SVneo cells. Additionally, a rat model of PE was generated by adenovirus injection via the tail vein to verify the role of OLFML3. RESULTS OLFML3 is highly expressed in both syncytiotrophoblasts and cytotrophoblasts and deregulated in preeclamptic placentas. OLFML3 overexpression in HTR-8/SVneo cells promoted cell proliferation, migration, invasion, and impeded apoptosis, and triggered phosphorylation on ser473 of AKT. Conversely, OLFML3 knockdown exerted opposite effects. Furthermore, OLFML3 overexpression ameliorates CoCl2-induced apoptosis of HTR-8/SVneo cells. In a rat model, OLFML3 overexpression alleviates PE-associated maternal symptoms, leading to lower blood pressure, less severe proteinuria, improved fetal growth restriction, as well as upregulation of P-AKT and downregulation of Cleaved caspase3 and Bax. DISCUSSION OLFML3 may alleviate PE development by inhibiting extravillous trophoblast cell apoptosis through the PI3K/AKT pathway. Our findings indicated that OLFML3 may provide a possible therapeutic target for PE.
Collapse
Affiliation(s)
- Haiying Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ruiping Li
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiangyujing Bian
- Pharmaceutical Research Lab, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoqing Li
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, 325000, China
| | - Cunjing Su
- Pharmaceutical Research Lab, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yang Wang
- Pharmaceutical Research Lab, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongping Zhang
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, 325000, China
| | - Jianqiong Zheng
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, 325000, China
| | - Yeping Wang
- Department of Obstetrics and Gynecology, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou Maternal and Child Health Care Hospital, Wenzhou, 325000, China
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China; Jiangsu Institute of Clinical Immunology, China.
| |
Collapse
|
14
|
Chen Z, Li Z, Zong Y, Xia B, Luo S, Deng G, Gao J. Exosome-delivered miR-410-3p reverses epithelial-mesenchymal transition, migration and invasion of trophoblasts in spontaneous abortion. J Cell Mol Med 2024; 28:e18097. [PMID: 38164738 PMCID: PMC10844701 DOI: 10.1111/jcmm.18097] [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: 05/14/2023] [Revised: 11/15/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
Current studies have indicated that insufficient trophoblast epithelial-mesenchymal transition (EMT), migration and invasion are crucial for spontaneous abortion (SA) occurrence and development. Exosomal miRNAs play significant roles in embryonic development and cellular communication. Hereon, we explored the roles of serum exosomes derived from SA patients on trophoblast EMT, migration and invasion. Exosomes were isolated from normal control (NC) patients with abortion for unplanned pregnancy and SA patients, then characterized by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and western blotting. Exosomal miRNA profiles were identified by miRNA sequencing. The effects of serum exosomes on trophoblast migration and invasion were detected by scratch wound healing and transwell assays, and other potential mechanisms were revealed by quantitative real-time PCR (RT-PCR), western blotting and dual-luciferase reporter assay. Finally, animal experiments were used to explore the effects of exosomal miR-410-3p on embryo absorption in mice. The serum exosomes from SA patients inhibited trophoblast EMT and reduced their migration and invasion ability in vitro. The miRNA sequencing showed that miR-410-3p was upregulated in SA serum exosomes. The functional experiments showed that SA serum exosomes restrained trophoblast EMT, migration and invasion by releasing miR-410-3p. Mechanistically, SA serum exosomal miR-410-3p inhibited trophoblast cell EMT, migration and invasion by targeting TNF receptor-associated factor 6 (TRAF6) at the post-transcriptional level. Besides, SA serum exosomal miR-410-3p inhibited the p38 MAPK signalling pathway by targeting TRAF6 in trophoblasts. Moreover, milk exosomes loaded with miR-410-3p mimic reached the maternal-fetal interface and aggravated embryo absorption in female mice. Clinically, miR-410-3p and TRAF6 expression were abnormal and negatively correlated in the placental villi of SA patients. Our findings indicated that exosome-derived miR-410-3p plays an important role between SA serum and trophoblasts in intercellular communication, suggesting a novel mechanism by which serum exosomal miRNA regulates trophoblasts in SA patients.
Collapse
Affiliation(s)
- Zhen‐yue Chen
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research Center of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Zhen Li
- The Second Clinical College of Guangzhou University of Chinese MedicineThe Second Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Yun Zong
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research Center of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Bo Xia
- The First Clinical Medical College of Guangzhou University of Chinese MedicineGuangzhouChina
- Lingnan Medical Research Center of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Song‐ping Luo
- Department of GynecologyFirst Affifiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Gao‐pi Deng
- Department of GynecologyFirst Affifiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Jie Gao
- Department of GynecologyFirst Affifiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
| |
Collapse
|
15
|
Wijdeven RH, Luk SJ, Schoufour TAW, van der Zanden SY, Cabezuelo M, Heemskerk MHM, Neefjes J. Balanced Epigenetic Regulation of MHC Class I Expression in Tumor Cells by the Histone Ubiquitin Modifiers BAP1 and PCGF1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:446-454. [PMID: 38088808 DOI: 10.4049/jimmunol.2300263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 11/13/2023] [Indexed: 01/18/2024]
Abstract
MHC class I (MHC-I) molecules are critical for CD8+ T cell responses to viral infections and malignant cells, and tumors can downregulate MHC-I expression to promote immune evasion. In this study, using a genome-wide CRISPR screen on a human melanoma cell line, we identified the polycomb repressive complex 1 (PRC1) subunit PCGF1 and the deubiquitinating enzyme BAP1 as opposite regulators of MHC-I transcription. PCGF1 facilitates deposition of ubiquitin at H2AK119 at the MHC-I promoters to silence MHC-I, whereas BAP1 removes this modification to restore MHC-I expression. PCGF1 is widely expressed in tumors and its depletion increased MHC-I expression in multiple tumor lines, including MHC-Ilow tumors. In cells characterized by poor MHC-I expression, PRC1 and PRC2 act in parallel to impinge low transcription. However, PCGF1 depletion was sufficient to increase MHC-I expression and restore T cell-mediated killing of the tumor cells. Taken together, our data provide an additional layer of regulation of MHC-I expression in tumors: epigenetic silencing by PRC1 subunit PCGF1.
Collapse
Affiliation(s)
- Ruud H Wijdeven
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, the Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam UMC, Amsterdam, the Netherlands
| | - Sietse J Luk
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tom A W Schoufour
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Sabina Y van der Zanden
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Marta Cabezuelo
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
16
|
Gao Y, Han W, Dong R, Wei S, Chen L, Gu Z, Liu Y, Guo W, Yan F. Efficient Reprogramming of Mouse Embryonic Stem Cells into Trophoblast Stem-like Cells via Lats Kinase Inhibition. BIOLOGY 2024; 13:71. [PMID: 38392290 PMCID: PMC10886645 DOI: 10.3390/biology13020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/24/2024]
Abstract
Mouse zygotes undergo multiple rounds of cell division, resulting in the formation of preimplantation blastocysts comprising three lineages: trophectoderm (TE), epiblast (EPI), and primitive endoderm (PrE). Cell fate determination plays a crucial role in establishing a healthy pregnancy. The initial separation of lineages gives rise to TE and inner cell mass (ICM), from which trophoblast stem cells (TSC) and embryonic stem cells (ESC) can be derived in vitro. Studying lineage differentiation is greatly facilitated by the clear functional distinction between TSC and ESC. However, transitioning between these two types of cells naturally poses challenges. In this study, we demonstrate that inhibiting LATS kinase promotes the conversion of ICM to TE and also effectively reprograms ESC into stable, self-renewing TS-like cells (TSLC). Compared to TSC, TSLC exhibits similar molecular properties, including the high expression of marker genes such as Cdx2, Eomes, and Tfap2c, as well as hypomethylation of their promoters. Importantly, TSLC not only displays the ability to differentiate into mature trophoblast cells in vitro but also participates in placenta formation in vivo. These findings highlight the efficient reprogramming of ESCs into TSLCs using a small molecular inducer, which provides a new reference for understanding the regulatory network between ESCs and TSCs.
Collapse
Affiliation(s)
- Yake Gao
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
- Reproductive Medicine Center, Wuhan Women's and Children's Medical Care Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenrui Han
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Rui Dong
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Shu Wei
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Lu Chen
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Zhaolei Gu
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yiming Liu
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Wei Guo
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Fang Yan
- State Key Laboratory of Conservation and Utilization of Bio-Resources, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| |
Collapse
|
17
|
Doria-Borrell P, Moya-Navamuel M, Hemberger M, Pérez-García V. Generation of Knockout Mouse Trophoblast Stem Cells by CRISPR/Cas9. Methods Mol Biol 2024; 2781:81-91. [PMID: 38502445 DOI: 10.1007/978-1-0716-3746-3_8] [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: 03/21/2024]
Abstract
The placenta is the organ that dictates the reproductive outcome of mammalian pregnancy by supplying nutrients and oxygen to the developing fetus to sustain its normal growth. During early mammalian development, trophoblast cells are the earliest cell type to differentiate with multipotent capacity to generate the trophoblast components of the placenta. The isolation and use of mouse trophoblast stem cells (mTSCs) to model in vitro trophoblast differentiation, in combination with CRISPR/Cas9 genome editing technology, has provided tremendous insight into the molecular mechanisms governing early mouse placentation. By knocking out a specific gene of interest in mTSCs, researchers are shedding light onto the molecular pathways involved in normal placental development and pregnancy disorders associated with abnormal placentation. In this chapter, we provide a detailed protocol for the genetic modification of mTSCs by using CRISPR/Cas9 genome editing system.
Collapse
Affiliation(s)
- Paula Doria-Borrell
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, Valencia, Spain
| | - Maria Moya-Navamuel
- Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, Valencia, Spain
| | - Myriam Hemberger
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | | |
Collapse
|
18
|
Pérez-García I, Pérez-García V. CRISPR Activation in Mouse Trophoblast Stem Cells. Methods Mol Biol 2024; 2781:93-103. [PMID: 38502446 DOI: 10.1007/978-1-0716-3746-3_9] [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: 03/21/2024]
Abstract
The placenta is a vital organ that regulates nutrient supply to the developing embryo during gestation. In mice, the placenta is composed of trophoblast lineage and mesodermal derivatives, which merge through the chorioallantoic fusion process in a critical event for the progression of placenta development. The trophoblast lineage is derived from self-renewing, multipotent cells known as mouse trophoblast stem cells (mTSCs). These cells are a valuable tool that allows scientists to comprehend the signals regulating major placental cell types' self-renewal and differentiation capacity. Recent advances in CRISPR-Cas9 genome editing applied in mTSCs have provided novel insights into the molecular networks involved in placentation. Here, we present a comprehensive CRISPR activation (CRISPRa) protocol based on the CRISPR/gRNA-directed synergistic activation mediator (SAM) method to overexpress specific target genes in mTSCs.
Collapse
|
19
|
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: 13] [Impact Index Per Article: 6.5] [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
|
20
|
Lin I, Wei A, Awamleh Z, Singh M, Ning A, Herrera A, Russell BE, Weksberg R, Arboleda VA. Multiomics of Bohring-Opitz syndrome truncating ASXL1 mutations identify canonical and noncanonical Wnt signaling dysregulation. JCI Insight 2023; 8:e167744. [PMID: 37053013 PMCID: PMC10322691 DOI: 10.1172/jci.insight.167744] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 04/07/2023] [Indexed: 04/14/2023] Open
Abstract
ASXL1 (additional sex combs-like 1) plays key roles in epigenetic regulation of early developmental gene expression. De novo protein-truncating mutations in ASXL1 cause Bohring-Opitz syndrome (BOS; OMIM #605039), a rare neurodevelopmental condition characterized by severe intellectual disabilities, distinctive facial features, hypertrichosis, increased risk of Wilms tumor, and variable congenital anomalies, including heart defects and severe skeletal defects giving rise to a typical BOS posture. These BOS-causing ASXL1 variants are also high-prevalence somatic driver mutations in acute myeloid leukemia. We used primary cells from individuals with BOS (n = 18) and controls (n = 49) to dissect gene regulatory changes caused by ASXL1 mutations using comprehensive multiomics assays for chromatin accessibility (ATAC-seq), DNA methylation, histone methylation binding, and transcriptome in peripheral blood and skin fibroblasts. Our data show that regardless of cell type, ASXL1 mutations drive strong cross-tissue effects that disrupt multiple layers of the epigenome. The data showed a broad activation of canonical Wnt signaling at the transcriptional and protein levels and upregulation of VANGL2, which encodes a planar cell polarity pathway protein that acts through noncanonical Wnt signaling to direct tissue patterning and cell migration. This multiomics approach identifies the core impact of ASXL1 mutations and therapeutic targets for BOS and myeloid leukemias.
Collapse
Affiliation(s)
- Isabella Lin
- Department of Human Genetics
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Department of Computational Medicine, UCLA, Los Angeles, California, USA
| | - Angela Wei
- Department of Human Genetics
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Department of Computational Medicine, UCLA, Los Angeles, California, USA
- Interdepartmental BioInformatics Program, UCLA, Los Angeles, California, USA
| | - Zain Awamleh
- Department of Genetics and Genome Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Meghna Singh
- Department of Human Genetics
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Department of Computational Medicine, UCLA, Los Angeles, California, USA
| | - Aileen Ning
- Department of Human Genetics
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Department of Computational Medicine, UCLA, Los Angeles, California, USA
| | - Analeyla Herrera
- Department of Human Genetics
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Department of Computational Medicine, UCLA, Los Angeles, California, USA
| | | | - Bianca E. Russell
- Division of Genetics, Department of Pediatrics, UCLA, Los Angeles, California, USA
| | - Rosanna Weksberg
- Department of Genetics and Genome Biology Program, Research Institute, The Hospital for Sick Children, Toronto, Ontario, Canada
- Division of Clinical & Metabolic Genetics, The Hospital for Sick Children, Toronto, Ontario, Canada
- Institute of Medical Sciences and Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Valerie A. Arboleda
- Department of Human Genetics
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
- Department of Computational Medicine, UCLA, Los Angeles, California, USA
- Interdepartmental BioInformatics Program, UCLA, Los Angeles, California, USA
- Molecular Biology Institute, UCLA, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA
| |
Collapse
|
21
|
Ursini G, Di Carlo P, Mukherjee S, Chen Q, Han S, Kim J, Deyssenroth M, Marsit CJ, Chen J, Hao K, Punzi G, Weinberger DR. Prioritization of potential causative genes for schizophrenia in placenta. Nat Commun 2023; 14:2613. [PMID: 37188697 PMCID: PMC10185564 DOI: 10.1038/s41467-023-38140-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 04/17/2023] [Indexed: 05/17/2023] Open
Abstract
Our earlier work has shown that genomic risk for schizophrenia converges with early life complications in affecting risk for the disorder and sex-biased neurodevelopmental trajectories. Here, we identify specific genes and potential mechanisms that, in placenta, may mediate such outcomes. We performed TWAS in healthy term placentae (N = 147) to derive candidate placental causal genes that we confirmed with SMR; to search for placenta and schizophrenia-specific associations, we performed an analogous analysis in fetal brain (N = 166) and additional placenta TWAS for other disorders/traits. The analyses in the whole sample and stratifying by sex ultimately highlight 139 placenta and schizophrenia-specific risk genes, many being sex-biased; the candidate molecular mechanisms converge on the nutrient-sensing capabilities of placenta and trophoblast invasiveness. These genes also implicate the Coronavirus-pathogenesis pathway and showed increased expression in placentae from a small sample of SARS-CoV-2-positive pregnancies. Investigating placental risk genes for schizophrenia and candidate mechanisms may lead to opportunities for prevention that would not be suggested by study of the brain alone.
Collapse
Affiliation(s)
- Gianluca Ursini
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Pasquale Di Carlo
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Group of Psychiatric Neuroscience, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Sreya Mukherjee
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Qiang Chen
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Shizhong Han
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Jiyoung Kim
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
| | - Maya Deyssenroth
- Departments of Environmental Medicine and Public Health, Icahn School of Public Health at Mount Sinai, New York, NY, USA
| | - Carmen J Marsit
- Departments of Environmental Health and Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Jia Chen
- Departments of Environmental Medicine and Public Health, Icahn School of Public Health at Mount Sinai, New York, NY, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giovanna Punzi
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, Johns Hopkins University Medical Campus, Baltimore, MD, USA.
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Seong J, Rivron NC. Protocol for capturing trophectoderm stem cells reflecting the blastocyst stage. STAR Protoc 2023; 4:102151. [PMID: 36930647 PMCID: PMC10031533 DOI: 10.1016/j.xpro.2023.102151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/13/2022] [Accepted: 02/10/2023] [Indexed: 03/18/2023] Open
Abstract
Classically, culturing mouse blastocysts with FGF4/TGF-β1, two epiblast-secreted inducers, allows for deriving trophoblast stem cells that comprise fluctuating subpopulations reflecting both pre- and post-implantation stages. However, a more complete combination of inducers (adding LPA, IL11, BMP7, Activin A, 8-Br-cAMP) captures trophectoderm stem cells with enhanced transcriptomic similarity to the blastocyst trophectoderm and self-renewal, reduced differentiation. Also, the complete combination of inducers increased potential to form blastoids and to instruct decidualization in utero, thus better reflecting the blastocyst. For complete details on the use and execution of this protocol, please refer to Seong et al.1.
Collapse
Affiliation(s)
- Jinwoo Seong
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, 1030 Vienna, Austria
| | - Nicolas C Rivron
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, 1030 Vienna, Austria.
| |
Collapse
|
23
|
Liu Z, He J, Jin P, Ran Y, Yin N, Qi H. CCL21/CCR7 Axis Contributes to Trophoblastic Cell Migration and Invasion in Preeclampsia by Affecting the Epithelial Mesenchymal Transition via the ERK1/2 Signaling Pathway. BIOLOGY 2023; 12:biology12020150. [PMID: 36829431 PMCID: PMC9952405 DOI: 10.3390/biology12020150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/20/2023]
Abstract
Preeclampsia (PE) is a pregnancy-related disorder that is a leading cause of maternal death. The failure of spiral artery remodeling due to insufficient trophoblast migration and invasion is critical in the pathogenesis of PE. Recently, the CC motif chemokine ligand 21 (CCL21) has been widely linked to cancer cell invasion and migration. However, their potential mechanisms are still unknown. In this study, we found that CCL21 expression was significantly lower in the PE group than that in the control group. In vitro experiments revealed that recombinant CCL21 could promote trophoblast cell epithelial-to-mesenchymal transitions (EMTs) and improve migration and invasion. Furthermore, an inhibitor of the ERK1/2 signaling pathway inhibited the CCL21-induced EMT process. Finally, a PE mouse model was established using the NOS inhibitor L-NAME, and we obtained similar results, with downregulated CCL21 and EMT biomarkers and upregulated CCR7. Taken together, these findings suggest that the CCL21/CCR7 axis influences EMT by activating the ERK1/2 signaling pathway, thereby affecting trophoblast cell migration and invasion, which may play a crucial role in the pathogenesis of PE.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jie He
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Pingsong Jin
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuxin Ran
- Department of Obstetrics, Women and Children’s Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Nanlin Yin
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
- Department of Center for Reproductive Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Correspondence: (N.Y.); (H.Q.)
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, Chongqing Medical University, Chongqing 400016, China
- Department of Obstetrics, Women and Children’s Hospital of Chongqing Medical University, Chongqing 401147, China
- Correspondence: (N.Y.); (H.Q.)
| |
Collapse
|
24
|
Li J, Qi Y, Yang K, Zhu L, Cui X, Liu Z. Follistatin Is a Novel Chemoattractant for Migration and Invasion of Placental Trophoblasts of Mice. Cells 2022; 11:cells11233816. [PMID: 36497076 PMCID: PMC9741044 DOI: 10.3390/cells11233816] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/19/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Follistatin (FST) as a gonadal protein is central to the establishment and maintenance of pregnancy. Trophoblasts' migration and invasion into the endometrium are critical events in placental development. This study aimed to elucidate the role of FST in the migration and invasion of placental trophoblasts of mice. We found that FST increased the vitality and proliferation of primary cultured trophoblasts of embryonic day 8.5 (E8.5) mice and promoted wound healing of trophoblasts. Moreover, FST significantly induced migration of trophoblasts in a microfluidic device and increased the number of invasive trophoblasts by Matrigel-coated transwell invasion assay. Being treated with FST, the adhesion of trophoblasts was inhibited, but intracellular calcium flux of trophoblasts was increased. Western blotting results showed that FST had no significant effects on the level of p-Smad3 or the ratio of p-Smad3/Smad3 in trophoblasts. Interestingly, FST elevated the level of p-JNK; the ratio of p-JNK/JNK; and expression of migration-related proteins N-cadherin, vimentin, ezrin and MMP2 in trophoblasts. Additionally, the migration of trophoblasts and expression of N-cadherin, vimentin, and MMP2 in trophoblasts induced by FST were attenuated by JNK inhibitor AS601245. These findings suggest that the elevated FST in pregnancy may act as a chemokine to induce trophoblast migration and invasion through the enhanced JNK signaling to maintain trophoblast function and promote placental development.
Collapse
Affiliation(s)
- Jing Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Yan Qi
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Key Laboratory of Neuroimmunology and Clinical Immunology, Changchun 130021, China
| | - Ke Yang
- Institute of Applied Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| | - Linjing Zhu
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xueling Cui
- Key Laboratory of Neuroimmunology and Clinical Immunology, Changchun 130021, China
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Zhonghui Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
- Key Laboratory of Neuroimmunology and Clinical Immunology, Changchun 130021, China
- Correspondence: ; Tel.: +86-431-8561-9476
| |
Collapse
|
25
|
Karvas RM, David L, Theunissen TW. Accessing the human trophoblast stem cell state from pluripotent and somatic cells. Cell Mol Life Sci 2022; 79:604. [PMID: 36434136 PMCID: PMC9702929 DOI: 10.1007/s00018-022-04549-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/01/2022] [Accepted: 09/05/2022] [Indexed: 11/26/2022]
Abstract
Trophoblasts are specialized epithelial cells that perform critical functions during blastocyst implantation and mediate maternal-fetal communication during pregnancy. However, our understanding of human trophoblast biology remains limited since access to first-trimester placental tissue is scarce, especially between the first and fourth weeks of development. Moreover, animal models inadequately recapitulate unique aspects of human placental physiology. In the mouse system, the isolation of self-renewing trophoblast stem cells has provided a valuable in vitro model system of placental development, but the derivation of analogous human trophoblast stem cells (hTSCs) has remained elusive until recently. Building on a landmark study reporting the isolation of bona fide hTSCs from blastocysts and first-trimester placental tissues in 2018, several groups have developed methods to derive hTSCs from pluripotent and somatic cell sources. Here we review the biological and molecular properties that define authentic hTSCs, the trophoblast potential of distinct pluripotent states, and methods for inducing hTSCs in somatic cells by direct reprogramming. The generation of hTSCs from pluripotent and somatic cells presents exciting opportunities to elucidate the molecular mechanisms of human placental development and the etiology of pregnancy-related diseases.
Collapse
Affiliation(s)
- Rowan M Karvas
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Laurent David
- Nantes Université, CHU Nantes, INSERM, CR2TI, UMR 1064, 44000, Nantes, France.
- Nantes Université, CHU Nantes, INSERM, CNRS, Biocore, US 016, UAR 3556, 44000, Nantes, France.
| | - Thorold W Theunissen
- Department of Developmental Biology and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
26
|
Padi2/3 Deficiency Alters the Epigenomic Landscape and Causes Premature Differentiation of Mouse Trophoblast Stem Cells. Cells 2022; 11:cells11162466. [PMID: 36010543 PMCID: PMC9406452 DOI: 10.3390/cells11162466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/02/2022] [Accepted: 08/06/2022] [Indexed: 11/17/2022] Open
Abstract
Histone citrullination is a relatively poorly studied epigenetic modification that involves the irreversible conversion of arginine residues into citrulline. It is conferred by a small family of enzymes known as protein arginine deiminases (PADIs). PADI function supports the pluripotent state of embryonic stem cells, but in other contexts, also promotes efficient cellular differentiation. In the current study, we sought to gain deeper insights into the possible roles of PADIs in mouse trophoblast stem cells (TSCs). We show that Padi2 and Padi3 are the most highly expressed PADI family members in TSCs and are rapidly down-regulated upon differentiation. Padi2/3 double knockout (DKO) TSCs express lower levels of stem cell transcription factors CDX2 and SOX2 and are prone to differentiate into extremely large trophoblast giant cells, an effect that may be mediated by centrosome duplication defects. Interestingly, Padi2/3 DKO TSCs display alterations to their epigenomic landscape, with fewer H3K9me3-marked chromocentric foci and globally reduced 5-methylcytosine levels. DNA methylation profiling identifies that this effect is specifically evident at CpG islands of critical trophoblast genes, such as Gata3, Peg3, Socs3 and Hand1. As a consequence of the hypomethylated state, these factors are up-regulated in Padi2/3 DKO TSCs, driving their premature differentiation. Our data uncover a critical epigenetic role for PADI2/3 in safeguarding the stem cell state of TSCs by modulating the DNA methylation landscape to restrict precocious trophoblast differentiation.
Collapse
|
27
|
Seong J, Frias-Aldeguer J, Holzmann V, Kagawa H, Sestini G, Heidari Khoei H, Scholte Op Reimer Y, Kip M, Pradhan SJ, Verwegen L, Vivié J, Li L, Alemany A, Korving J, Darmis F, van Oudenaarden A, Ten Berge D, Geijsen N, Rivron NC. Epiblast inducers capture mouse trophectoderm stem cells in vitro and pattern blastoids for implantation in utero. Cell Stem Cell 2022; 29:1102-1118.e8. [PMID: 35803228 DOI: 10.1016/j.stem.2022.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/21/2022] [Accepted: 06/02/2022] [Indexed: 11/03/2022]
Abstract
The embryo instructs the allocation of cell states to spatially regulate functions. In the blastocyst, patterning of trophoblast (TR) cells ensures successful implantation and placental development. Here, we defined an optimal set of molecules secreted by the epiblast (inducers) that captures in vitro stable, highly self-renewing mouse trophectoderm stem cells (TESCs) resembling the blastocyst stage. When exposed to suboptimal inducers, these stem cells fluctuate to form interconvertible subpopulations with reduced self-renewal and facilitated differentiation, resembling peri-implantation cells, known as TR stem cells (TSCs). TESCs have enhanced capacity to form blastoids that implant more efficiently in utero due to inducers maintaining not only local TR proliferation and self-renewal, but also WNT6/7B secretion that stimulates uterine decidualization. Overall, the epiblast maintains sustained growth and decidualization potential of abutting TR cells, while, as known, distancing imposed by the blastocyst cavity differentiates TR cells for uterus adhesion, thus patterning the essential functions of implantation.
Collapse
Affiliation(s)
- Jinwoo Seong
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Javier Frias-Aldeguer
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands; Maastricht University, Maastricht, the Netherlands
| | - Viktoria Holzmann
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Harunobu Kagawa
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Giovanni Sestini
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Heidar Heidari Khoei
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Yvonne Scholte Op Reimer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Maarten Kip
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | - Saurabh J Pradhan
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria
| | - Lucas Verwegen
- Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Judith Vivié
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | - Linfeng Li
- Maastricht University, Maastricht, the Netherlands
| | - Anna Alemany
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | - Jeroen Korving
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | - Frank Darmis
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands
| | | | - Derk Ten Berge
- Department of Cell Biology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Niels Geijsen
- Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands; Department of Anatomy and Embryology, LUMC, Leiden University, Leiden, the Netherlands
| | - Nicolas C Rivron
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna Biocenter, Vienna, Austria; Hubrecht Institute for Developmental Biology and Stem Cell Research, Utrecht, the Netherlands; Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
28
|
Sheridan MA, Zhao X, Fernando RC, Gardner L, Perez-Garcia V, Li Q, Marsh SGE, Hamilton R, Moffett A, Turco MY. Characterization of primary models of human trophoblast. Development 2021; 148:272500. [PMID: 34651188 PMCID: PMC8602945 DOI: 10.1242/dev.199749] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023]
Abstract
Two recently developed models, trophoblast organoids and trophoblast stem cells (TSCs), are useful tools to further the understanding of human placental development. Both differentiate from villous cytotrophoblast (VCT) to either extravillous trophoblast (EVT) or syncytiotrophoblast (SCT). Here, we compare the transcriptomes and miRNA profiles of these models to identify which trophoblast they resemble in vivo. Our findings indicate that TSCs do not readily undergo SCT differentiation and closely resemble cells at the base of the cell columns from where EVT derives. In contrast, organoids are similar to VCT and undergo spontaneous SCT differentiation. A defining feature of human trophoblast is that VCT and SCT are human leukocyte antigen (HLA) null, whereas EVT expresses HLA-C, -G and -E molecules. We find that trophoblast organoids retain these in vivo characteristics. In contrast, TSCs express classical HLA-A and HLA-B molecules, and maintain their expression after EVT differentiation, with upregulation of HLA-G. Furthermore, HLA expression in TSCs differs when grown in 3D rather than in 2D, suggesting that mechanical cues are important. Our results can be used to select the most suitable model for the study of trophoblast development, function and pathology. Summary: Characterization of trophoblast organoids and trophoblast stem cells as exciting models of human placentation enables the selection of the most suitable system to address specific research questions.
Collapse
Affiliation(s)
- Megan A Sheridan
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Xiaohui Zhao
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.,Department of Physiology, Neuroscience and Development, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ridma C Fernando
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Lucy Gardner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Vicente Perez-Garcia
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.,Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, Valencia 46012, Spain
| | - Qian Li
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Steven G E Marsh
- Anthony Nolan Research Institute, Royal Free Hospital, London NW3 2QG, UK.,UCL Cancer Institute, Royal Free Campus, London WC1E 6DD, UK
| | - Russell Hamilton
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.,Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Margherita Y Turco
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK.,Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|