1
|
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
|
2
|
Forman-Rubinsky R, Feng W, Schlegel BT, Paul A, Zuppo D, Kedziora K, Stoltz D, Watkins S, Rajasundaram D, Li G, Tsang M. Cited4a limits cardiomyocyte dedifferentiation and proliferation during zebrafish heart regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.626917. [PMID: 39713454 PMCID: PMC11661073 DOI: 10.1101/2024.12.05.626917] [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
Cardiac regeneration involves the interplay of complex interactions between many different cell types, including cardiomyocytes. The exact mechanism that enables cardiomyocytes to undergo dedifferentiation and proliferation to replace lost cells has been intensely studied. Here we report a single nuclear RNA sequencing profile of the injured zebrafish heart and identify distinct cardiomyocyte populations in the injured heart. These cardiomyocyte populations have diverse functions, including stress response, myofibril assembly, proliferation and contraction. The contracting cardiomyocyte population also involves the activation of maturation pathways as an early response to injury. This intriguing finding suggests that constant maintenance of a distinctive terminally differentiated cardiomyocyte population is important for cardiac function during regeneration. To test this hypothesis, we determined that cited4a, a p300/CBP transcriptional coactivator, is induced after injury in the mature cardiomyocyte population. Moreover, loss-of-cited4a mutants presented increased dedifferentiation, proliferation and accelerated heart regeneration. Thus, suppressing cardiomyocyte maturation pathway activity in injured hearts could be an approach to promote heart regeneration.
Collapse
Affiliation(s)
- Rachel Forman-Rubinsky
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Wei Feng
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Brent T Schlegel
- Department of Pediatrics, Division of Health Informatics, Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Angela Paul
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Daniel Zuppo
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Current address: Department of Medicine, University of Rochester Medical Center Rochester, NY
| | - Katarzyna Kedziora
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Donna Stoltz
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Simon Watkins
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Biological Imaging, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Division of Health Informatics, Children's Hospital of Pittsburgh, Pittsburgh, PA
| | - Guang Li
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Michael Tsang
- Department of Cell Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
- Center for Integrative Organ Systems, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| |
Collapse
|
3
|
Aşır F, Özalp Z, Yülek ÖU, Erdemci F, Korak T, Taş F. CITED1 expression in odontogenic cysts. BMC Oral Health 2024; 24:782. [PMID: 38997708 PMCID: PMC11242007 DOI: 10.1186/s12903-024-04413-4] [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: 03/06/2024] [Accepted: 05/27/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Originating from odontogenic tissue, Odontogenic cysts are pathological cavities lined with epithelial cells and surrounded by fibrous connective tissue. This study investigated expression of CITED1 protein in different types of odontogenic cysts. MATERIAL AND METHOD 40 keratocysts, 40 radicular cysts, and 40 dentigerous cysts were excised and processed for routine paraffin wax embedding protocol. Macroscopic and panoramic radiographies images were used for diagnosis. Demographical properties and dental parameters were recorded. Cystic tissues were stained with hematoxylin-eosin dye and CITED1 antibody. Semi-quantitative analysis was performed for immune staining. The protein-protein interaction network, hub gene detection and KEGG analysis were conducted using Cytoscape software. RESULT Odontogenic keratocysts was imaged with 6-8 layered epithelial cells and fibrous cyst walls with inflammatory cells. Radicular cysts had stratified squamous epithelium with varying thickness, ciliated cells, and Rushton hyaline bodies. Dentigerous cysts presented hyperplastic non-keratinized epithelium, fibrous tissue, rete ridges, and inflammatory cells. CITED1 immunoexpression was highest in odontogenic keratocysts, followed by radicular cysts, and lowest in dentigerous cysts. Nuclear and cytoplasmic CITED1 expression was significantly elevated in odontogenic keratocysts compared to radicular and dentigerous cysts. The top five targets of CITED1 were identified, primarily showing enrichment in hormone and cancer related pathways. CONCLUSIONS Positive CITED1 expression in all three types of odontogenic cysts suggest a potential role for CITED1 in the pathogenesis of odontogenic cysts, particularly in keratocysts. Further investigations are needed to elucidate the exact mechanisms underlying the differential expression of CITED1 and its implications for the development and progression of odontogenic cysts.
Collapse
Affiliation(s)
- Fırat Aşır
- Department of Histology and Embryology, Medical Faculty, Dicle University, Diyarbakır, Turkey.
| | - Zeki Özalp
- Department of Oral, Dental and Maxillofacial Surgery, Siirt Oral and Dental Health Center, Siirt, Turkey
| | - Özden Uçtu Yülek
- Department of Pathology, Çanakkale 18 Mart University, Çanakkale, Turkey
| | - Fikri Erdemci
- Department of Histology and Embryology, Medical Faculty, Dicle University, Diyarbakır, Turkey
| | - Tugcan Korak
- Department of Medical Biology, Medical Faculty, Kocaeli University, Kocaeli, Turkey
| | - Fatih Taş
- Department of Histology and Embryology, Medical Faculty, Siirt University, Siirt, Turkey
| |
Collapse
|
4
|
Yang J, Bao Z, Li J, Lu T, Cai J, Sun S, Shen N, Chen Y, Zhao B, Wu X. MTHFR as a Novel Candidate Marker for Litter Size in Rabbits. Animals (Basel) 2024; 14:1930. [PMID: 38998044 PMCID: PMC11240429 DOI: 10.3390/ani14131930] [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: 03/22/2024] [Revised: 06/15/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Litter size is a significant economic trait during animal reproduction. This current study attempted to decipher whether MTHFR promotes the apoptosis of granulosa cells (GCs) and inhibits their proliferation by investigating the effects of the MTHFR gene using flow cytometry and a Cell Counting Kit-8 (CCK-8) assay. MTHFR is linked with ovarian follicle development in the reproductive performance of 104 female New Zealand rabbits. We observed that MTHFR could regulate the mRNA of follicular development-related genes (TIMP1, CITED1, FSHR, GHR, HSD17B1, and STAR) with a qRT-PCR, and we observed the protein expression of CITED1 and GHR using a western blot (WB) analysis. The dual luciferase activity assays helped identify the core promoter region of the MTHFR gene, and the polymorphism of the MTHFR promoter region was studied using Sanger sequencing. The results indicated four single nucleotide polymorphisms (SNPs) within the core promoter region, among which the g.-680C>A locus was significantly associated with both the total and alive litter sizes. Additionally, the CC genotype was associated with the largest total and alive litter sizes, compared to the CA and AA genotypes (p < 0.05). In conclusion, this study investigated the effects of MTHFR on ovarian granulosa cells and its association with selected reproductive parameters in rabbits. The results provide a theoretical foundation for the use of MTHFR as a molecular marker in rabbits.
Collapse
Affiliation(s)
- Jie Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Zhiyuan Bao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jiali Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Tingting Lu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jiawei Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Shaoning Sun
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Ning Shen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yang Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Bohao Zhao
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Xinsheng Wu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture & Agri-Product Safety, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
5
|
Vogtmann R, Riedel A, Sassmannshausen I, Langer S, Kühnel-Terjung E, Kimmig R, Schorle H, Winterhager E, Gellhaus A. Overexpression of Human sFLT1 in the Spongiotrophoblast Is Sufficient to Induce Placental Dysfunction and Fetal Growth Restriction in Transgenic Mice. Int J Mol Sci 2024; 25:2040. [PMID: 38396719 PMCID: PMC10888837 DOI: 10.3390/ijms25042040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Preeclampsia (PE) is characterized by maternal hypertension and placental dysfunction, often leading to fetal growth restriction (FGR). It is associated with an overexpression of the anti-angiogenic sFLT1 protein, which originates from the placenta and serves as a clinical biomarker to predict PE. To analyze the impact of sFLT1 on placental function and fetal growth, we generated transgenic mice with placenta-specific human sFLT1 (hsFLT1) overexpression. Immunohistochemical, morphometrical, and molecular analyses of the placentas on 14.5 dpc and 18.5 dpc were performed with a focus on angiogenesis, nutrient transport, and inflammation. Additionally, fetal development upon placental hsFLT1 overexpression was investigated. Dams exhibited a mild increase in serum hsFLT1 levels upon placental hsFLT1 expression and revealed growth restriction of the fetuses in a sex-specific manner. Male FGR fetuses expressed higher amounts of placental hsFLT1 mRNA compared to females. FGR placentas displayed an altered morphology, hallmarked by an increase in the spongiotrophoblast layer and changes in labyrinthine vascularization. Further, FGR placentas showed a significant reduction in placental glycogen storage and nutrient transporter expression. Moreover, signs of hypoxia and inflammation were observed in FGR placentas. The transgenic spongiotrophoblast-specific hsFLT1 mouse line demonstrates that low hsFLT1 serum levels are sufficient to induce significant alterations in fetal and placental development in a sex-specific manner.
Collapse
Affiliation(s)
- Rebekka Vogtmann
- Department of Gynecology and Obstetrics, University Hospital, 45147 Essen, Germany (A.R.); (R.K.)
| | - Alina Riedel
- Department of Gynecology and Obstetrics, University Hospital, 45147 Essen, Germany (A.R.); (R.K.)
| | - Ivanka Sassmannshausen
- Department of Gynecology and Obstetrics, University Hospital, 45147 Essen, Germany (A.R.); (R.K.)
| | - Sarah Langer
- Department of Gynecology and Obstetrics, University Hospital, 45147 Essen, Germany (A.R.); (R.K.)
| | - Elisabeth Kühnel-Terjung
- Department of Gynecology and Obstetrics, University Hospital, 45147 Essen, Germany (A.R.); (R.K.)
| | - Rainer Kimmig
- Department of Gynecology and Obstetrics, University Hospital, 45147 Essen, Germany (A.R.); (R.K.)
| | - Hubert Schorle
- Department of Developmental Pathology, Institute of Pathology, University Hospital Bonn, 53127 Bonn, Germany;
| | - Elke Winterhager
- EM Unit, Imaging Center Essen, University Hospital, 45147 Essen, Germany;
| | - Alexandra Gellhaus
- Department of Gynecology and Obstetrics, University Hospital, 45147 Essen, Germany (A.R.); (R.K.)
| |
Collapse
|
6
|
Chen W, Jiang J, Gao J, Wang G, Wang R, Lv J, Ben J. Roles and signaling pathways of CITED1 in tumors: overview and novel insights. J Int Med Res 2024; 52:3000605231220890. [PMID: 38190845 PMCID: PMC10775745 DOI: 10.1177/03000605231220890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024] Open
Abstract
CBP/p300 interacting transactivator with Glu/Asp-rich carboxy-terminal domain 1 (CITED1) is a transcriptional activator belonging to the non-DNA-binding transcription co-regulator family. It regulates diverse pathways, including the transforming growth factor/bone morphogenetic protein/SMAD, estrogen, Wnt-β-catenin, and androgen-AR signaling pathways, by binding to CBP/p300 co-activators through its conserved transactivation domain CR2. CITED1 plays an important role in embryonic development and a certain regulatory role in the occurrence and development of various tumors. In this article, the biological characteristics, expression regulation, participating signaling pathways, and potential roles of CITED1 in the clinical diagnosis and treatment of tumors are reviewed.
Collapse
Affiliation(s)
- Wenting Chen
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, China
| | - Jianing Jiang
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, China
| | - Jinqi Gao
- Department of Intervention, The Second Hospital Affiliated to Dalian Medical University, Dalian, China
| | - Gang Wang
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ruoyu Wang
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
- The Key Laboratory of Biomarker High Throughput Screening and Target Translation of Breast and Gastrointestinal Tumor, Dalian, China
| | - Jinyan Lv
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Jing Ben
- Department of Oncology Medicine, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| |
Collapse
|
7
|
Jeong DS, Lee JY, Kim MH, Oh JH. Regulation of sexually dimorphic placental adaptation in LPS exposure-induced intrauterine growth restriction. Mol Med 2023; 29:114. [PMID: 37718409 PMCID: PMC10506314 DOI: 10.1186/s10020-023-00688-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 06/15/2023] [Indexed: 09/19/2023] Open
Abstract
BACKGROUND Sexual dimorphism in placental physiology affects the functionality of placental adaptation during adverse pregnancy. Defects of placental function compromise fetal programming, affecting the offspring's adult life. However, studies focusing on the relationship between sex-specific placental adaptation and consequent fetal maldevelopment under sub-optimal uterus milieu are still elusive. METHODS Here, we investigated the effects of maternal lipopolysaccharide (LPS) exposure between placental sex. Pregnant ICR mice received intraperitoneal injection of phosphate-buffered saline or 100, 200, and 400 µg/kg LPS on the gestational day (GD) 15.5. To determine whether prenatal maternal LPS exposure resulted in complicated pregnancy outcomes, survival rate of embryos was calculated and the growth of embryos and placentas was examined. To elucidate global transcriptomic changes occurring in the placenta, total RNA-sequencing (RNA-seq) was performed in female and male placentas. RESULTS LPS administration induced placental inflammation in both sexes at GD 17.5. Prenatal infection resulted in growth retardation in both sexes of embryos, and especially more prevalently in male. Impaired placental development was observed in a sex-specific manner. LPS 400 µg/kg reduced the percentage area of the labyrinth in females and junctional zone in males, respectively. RNA-sequencing revealed widespread sexually dimorphic transcriptional changes in placenta. In particular, representative changes were involved in biological processes such as trophoblast differentiation, nutrient/ion transporter, pregnancy, and immune system. CONCLUSIONS Our results present the sexually dimorphic responses of placental physiology in intrauterine growth restriction model and provide tentative relationship further to be elucidated between sex-biased placental functional change and long-term effects on the offspring's later life.
Collapse
Affiliation(s)
- Da Som Jeong
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Ji-Yeon Lee
- Vivozon, Inc, Kolon Digital Tower3, 49, Achasan-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Myoung Hee Kim
- Department of Anatomy, Embryology Laboratory, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Ji Hoon Oh
- Department of Biological Sciences, Keimyung University College of Natural Sciences, Daegu, 42601, Republic of Korea.
| |
Collapse
|
8
|
Cuny H, Bozon K, Kirk RB, Sheng DZ, Bröer S, Dunwoodie SL. Maternal heterozygosity of Slc6a19 causes metabolic perturbation and congenital NAD deficiency disorder in mice. Dis Model Mech 2023; 16:282089. [PMID: 36374036 PMCID: PMC9702539 DOI: 10.1242/dmm.049647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/06/2022] [Indexed: 11/16/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD) is a key metabolite synthesised from vitamin B3 or tryptophan. Disruption of genes encoding NAD synthesis enzymes reduces NAD levels and causes congenital NAD deficiency disorder (CNDD), characterised by multiple congenital malformations. SLC6A19 (encoding B0AT1, a neutral amino acid transporter), represents the main transporter for free tryptophan in the intestine and kidney. Here, we tested whether Slc6a19 heterozygosity in mice limits the tryptophan available for NAD synthesis during pregnancy and causes adverse pregnancy outcomes. Pregnant Slc6a19+/- mice were fed diets depleted of vitamin B3, so that tryptophan was the source of NAD during gestation. This perturbed the NAD metabolome in pregnant Slc6a19+/- females, resulting in reduced NAD levels and increased rates of embryo loss. Surviving embryos were small and exhibited specific combinations of CNDD-associated malformations. Our results show that genes not directly involved in NAD synthesis can affect NAD metabolism and cause CNDD. They also suggest that human female carriers of a SLC6A19 loss-of-function allele might be susceptible to adverse pregnancy outcomes unless sufficient NAD precursor amounts are available during gestation. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Hartmut Cuny
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, Sydney, NSW 2052, Australia
| | - Kayleigh Bozon
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
| | - Rosemary B. Kirk
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
| | - Delicia Z. Sheng
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
| | - Stefan Bröer
- Research School of Biology, Australian National University, Canberra, ACT 0200, Australia
| | - Sally L. Dunwoodie
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Sydney, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, Sydney, NSW 2052, Australia
- Faculty of Science, University of New South Wales, Sydney, NSW 2052, Australia
- Author for correspondence ()
| |
Collapse
|
9
|
Abstract
Establishment of the hemochorial uterine-placental interface requires exodus of trophoblast cells from the placenta and their transformative actions on the uterus, which represent processes critical for a successful pregnancy, but are poorly understood. We examined the involvement of CBP/p300-interacting transactivator with glutamic acid/aspartic acid-rich carboxyl-terminal domain 2 (CITED2) in rat and human trophoblast cell development. The rat and human exhibit deep hemochorial placentation. CITED2 was distinctively expressed in the junctional zone (JZ) and invasive trophoblast cells of the rat. Homozygous Cited2 gene deletion resulted in placental and fetal growth restriction. Small Cited2 null placentas were characterized by disruptions in the JZ, delays in intrauterine trophoblast cell invasion, and compromised plasticity. In the human placentation site, CITED2 was uniquely expressed in the extravillous trophoblast (EVT) cell column and importantly contributed to the development of the EVT cell lineage. We conclude that CITED2 is a conserved regulator of deep hemochorial placentation.
Collapse
|
10
|
Ortega MS, Rizo JA, Drum JN, O'Neil EV, Pohler KG, Kerns K, Schmelze A, Green J, Spencer TE. Development of an Improved in vitro Model of Bovine Trophectoderm Differentiation. FRONTIERS IN ANIMAL SCIENCE 2022. [DOI: 10.3389/fanim.2022.898808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mechanisms regulating early stages of placentation and trophectoderm differentiation in the ruminant conceptus remain poorly understood. Here we present a model of trophectoderm (TE) differentiation in vitro from outgrowths of individual in vitro derived embryos. Cell outgrowths expressed markers of mononucleate (MNC) and binucleate (BNC) TE cells. The percentage of BNC ranged from 14 to 39% in individual outgrowths as determined by flow cytometry. Pregnancy-associated glycoproteins (PAGs), produced by BNC, were measured in culture media on days 35 to 54. Continuous secretion of PAGs was observed and indicative of BNC functionality. Gene expression was evaluated in 20 embryo cell outgrowths derived from two different sires. Expression of HAND1, which is involved in TE differentiation, and CSH2, a BNC-specific gene, was altered in cell outgrowths between the two sires tested. Single-cell RNA-seq analysis of day 40 TE cell outgrowths revealed 11 distinct cell populations, with specific clusters genes involved in TE lineage specification, proliferation, and differentiation. In addition, whole -RNAseq analysis was performed in day 35 and 40 TE cell outgrowths and confirmed sustained expression of genes expressed by BNC, such as CSH2 and some PAGs. The developed in vitro bovine embryo outgrowth culture found evidence for MNC and BNC differentiation and continuous production of PAGs, recapitulating key features of early bovine placenta development. This model can be used to understand the developmental biology of TE cells, provide insights into paternal influences on TE differentiation, and impact our understanding of early pregnancy loss in cattle.
Collapse
|
11
|
Elmore SA, Cochran RZ, Bolon B, Lubeck B, Mahler B, Sabio D, Ward JM. Histology Atlas of the Developing Mouse Placenta. Toxicol Pathol 2021; 50:60-117. [PMID: 34872401 PMCID: PMC8678285 DOI: 10.1177/01926233211042270] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The use of the mouse as a model organism is common in translational research. This mouse-human similarity holds true for placental development as well. Proper formation of the placenta is vital for development and survival of the maturing embryo. Placentation involves sequential steps with both embryonic and maternal cell lineages playing important roles. The first step in placental development is formation of the blastocyst wall (approximate embryonic days [E] 3.0-3.5). After implantation (∼E4.5), extraembryonic endoderm progressively lines the inner surface of the blastocyst wall (∼E4.5-5.0), forming the yolk sac that provides histiotrophic support to the embryo; subsequently, formation of the umbilical vessels (∼E8.5) supports transition to the chorioallantoic placenta and hemotrophic nutrition. The fully mature ("definitive") placenta is established by ∼E12.5. Abnormal placental development often leads to embryonic mortality, with the timing of death depending on when placental insufficiency takes place and which cells are involved. This comprehensive macroscopic and microscopic atlas highlights the key features of normal and abnormal mouse placental development from E4.5 to E18.5. This in-depth overview of a transient (and thus seldom-analyzed) developmental tissue should serve as a useful reference to aid researchers in identifying and describing mouse placental changes in engineered, induced, and spontaneous disease models.
Collapse
Affiliation(s)
- Susan A Elmore
- National Toxicology Program, 6857National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Robert Z Cochran
- National Toxicology Program, 6857National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | - Beth Lubeck
- National Toxicology Program, 6857National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Beth Mahler
- Experimental Pathology Laboratories, Inc., Research Triangle Park, NC, USA
| | - David Sabio
- Experimental Pathology Laboratories, Inc., Research Triangle Park, NC, USA
| | - Jerrold M Ward
- Global Vet Pathology, Montgomery Village, MD, USA *Co-first authors
| |
Collapse
|
12
|
Torregrosa-Carrión R, Piñeiro-Sabarís R, Siguero-Álvarez M, Grego-Bessa J, Luna-Zurita L, Fernandes VS, MacGrogan D, Stainier DYR, de la Pompa JL. Adhesion G protein-coupled receptor Gpr126/Adgrg6 is essential for placental development. SCIENCE ADVANCES 2021; 7:eabj5445. [PMID: 34767447 PMCID: PMC8589310 DOI: 10.1126/sciadv.abj5445] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Mutations in the G protein–coupled receptor GPR126/ADGRG6 cause human diseases, including defective peripheral nervous system (PNS) myelination. To study GPR126 function, we generated new genetic mice and zebrafish models. Murine Gpr126 is expressed in developing heart endocardium, and global Gpr126 inactivation is embryonically lethal, with mutants having thin-walled ventricles but unaffected heart patterning or maturation. Endocardial-specific Gpr126 deletion does not affect heart development or function, and transgenic endocardial GPR126 expression fails to rescue lethality in Gpr126-null mice. Zebrafish gpr126 mutants display unaffected heart development. Gpr126 is also expressed in placental trophoblast giant cells. Gpr126-null mice with a heterozygous placenta survive but exhibit GPR126-defective PNS phenotype. In contrast, Gpr126-null embryos with homozygous mutant placenta die but are rescued by placental GPR126 expression. Gpr126-deficient placentas display down-regulation of preeclampsia markers Mmp9, Cts7, and Cts8. We propose that the placenta-heart axis accounts for heart abnormalities secondary to placental defects in Gpr126 mutants.
Collapse
Affiliation(s)
- Rebeca Torregrosa-Carrión
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Rebeca Piñeiro-Sabarís
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Marcos Siguero-Álvarez
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Joaquím Grego-Bessa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Luis Luna-Zurita
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Vitor Samuel Fernandes
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
- Corresponding author.
| |
Collapse
|
13
|
Mateo-Otero Y, Sánchez JM, Recuero S, Bagés-Arnal S, McDonald M, Kenny DA, Yeste M, Lonergan P, Fernandez-Fuertes B. Effect of Exposure to Seminal Plasma Through Natural Mating in Cattle on Conceptus Length and Gene Expression. Front Cell Dev Biol 2020; 8:341. [PMID: 32478076 PMCID: PMC7235327 DOI: 10.3389/fcell.2020.00341] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022] Open
Abstract
A growing body of evidence suggests that paternal factors have an impact on offspring development. These studies have been mainly carried out in mice, where seminal plasma (SP) has been shown to regulate endometrial gene expression and impact embryo development and subsequent offspring health. In cattle, infusion of SP into the uterus also induces changes in endometrial gene expression, however, evidence for an effect of SP on early embryo development is lacking. In addition, during natural mating, the bull ejaculates in the vagina; hence, it is not clear whether any SP reaches the uterus in this species. Thus, the aim of the present study was to determine whether SP exposure leads to improved early embryo survival and developmental rates in cattle. To this end, Day 7 in vitro produced blastocysts were transferred to heifers (12-15 per heifer) previously mated to vasectomized bulls (n = 13 heifers) or left unmated (n = 12 heifers; control). At Day 14, heifers were slaughtered, and conceptuses were recovered to assess size, morphology and expression of candidate genes involved in different developmental pathways. Additionally, CL volume at Day 7, and weight and volume of CL at Day 14 were recorded. No effect of SP on CL volume and weight not on conceptus recovery rate was observed. However, filamentous conceptuses recovered from SP-exposed heifers were longer in comparison to the control group and differed in expression of CALM1, CITED1, DLD, HNRNPDL, PTGS2, and TGFB3. In conclusion, data indicate that female exposure to SP during natural mating can affect conceptus development in cattle. This is probably achieved through modulation of the female reproductive environment at the time of mating.
Collapse
Affiliation(s)
- Yentel Mateo-Otero
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
| | - José María Sánchez
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Sandra Recuero
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
| | - Sandra Bagés-Arnal
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Michael McDonald
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - David A. Kenny
- Animal and Bioscience Research Department, Animal and Grassland Research and Innovation Centre, Teagasc Grange, Dunsany, Ireland
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
| | - Pat Lonergan
- School of Agriculture and Food Science, University College Dublin, Dublin, Ireland
| | - Beatriz Fernandez-Fuertes
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Institute of Food and Agricultural Technology, University of Girona, Girona, Spain
- Unit of Cell Biology, Department of Biology, Faculty of Sciences, University of Girona, Girona, Spain
| |
Collapse
|
14
|
Li H, Song M, Yang W, Cao P, Zheng L, Zuo Y. A Comparative Analysis of Single-Cell Transcriptome Identifies Reprogramming Driver Factors for Efficiency Improvement. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1053-1064. [PMID: 32045876 PMCID: PMC7015826 DOI: 10.1016/j.omtn.2019.12.035] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/23/2019] [Accepted: 12/26/2019] [Indexed: 12/11/2022]
Abstract
Terminally differentiated somatic cells can be reprogrammed into a totipotent state through somatic cell nuclear transfer (SCNT). The incomplete reprogramming is the major reason for developmental arrest of SCNT embryos at early stages. In our studies, we found that pathways for autophagy, endocytosis, and apoptosis were incompletely activated in nuclear transfer (NT) 2-cell arrest embryos, whereas extensively inhibited pathways for stem cell pluripotency maintenance, DNA repair, cell cycle, and autophagy may result in NT 4-cell embryos arrest. As for NT normal embryos, a significant shift in expression of developmental transcription factors (TFs) Id1, Pou6f1, Cited1, and Zscan4c was observed. Compared with pluripotent gene Ascl2 being activated only in NT 2-cell, Nanog, Dppa2, and Sall4 had major expression waves in normal development of both NT 2-cell and 4-cell embryos. Additionally, Kdm4b/4d and Kdm5b had been confirmed as key markers in NT 2-cell and 4-cell embryos, respectively. Histone acetylases Kat8, Elp6, and Eid1 were co-activated in NT 2-cell and 4-cell embryos to facilitate normal development. Gadd45a as a key driver functions with Tet1 and Tet2 to improve the efficiency of NT reprogramming. Taken together, our findings provided an important theoretical basis for elucidating the potential molecular mechanisms and identified reprogramming driver factor to improve the efficiency of SCNT reprogramming.
Collapse
Affiliation(s)
- Hanshuang Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Mingmin Song
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Wuritu Yang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Pengbo Cao
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Lei Zheng
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China
| | - Yongchun Zuo
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot 010070, China.
| |
Collapse
|
15
|
Ortega MS, Moraes JGN, Patterson DJ, Smith MF, Behura SK, Poock S, Spencer TE. Influences of sire conception rate on pregnancy establishment in dairy cattle. Biol Reprod 2019; 99:1244-1254. [PMID: 29931362 DOI: 10.1093/biolre/ioy141] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022] Open
Abstract
Establishment of pregnancy in cattle is complex and encompasses ovulation, fertilization, blastocyst formation and growth into an elongated conceptus, pregnancy recognition signaling, and development of the embryo and placenta. The objective here was to investigate sire influences on pregnancy establishment in cattle. First, 10 Holstein bulls were classified as high or low fertility based on their sire conception rate (SCR) value. In a field trial, pregnancy at first timed insemination was not different between high and low SCR bulls. Next, 5 of the 10 sires were phenotyped using in vitro and in vivo embryo production. There was no effect of SCR classification on in vitro embryo cleavage rate, but low SCR sires produced fewer day 8 blastocysts. In superovulated heifers, high SCR bulls produced a lower percentage of unfertilized oocytes and fewer degenerated embryos compared to low SCR bulls. Recipient heifers received three to five in vivo produced embryos from either high or low SCR sires on day 7 postestrus. Day 16 conceptus recovery and length were not different between SCR groups, and the conceptus transcriptome was not appreciably different between high and low SCR sires. The reduced ability of embryos from low SCR bulls to establish pregnancy is multifactorial and encompasses sperm fertilizing ability, preimplantation embryonic development, and development of the embryo and placenta after conceptus elongation and pregnancy recognition. These studies highlight the importance of understanding genetic contributions of the sire to pregnancy establishment that is crucial to increase reproductive efficiency in dairy cattle.
Collapse
Affiliation(s)
- M Sofia Ortega
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - João G N Moraes
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - David J Patterson
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Michael F Smith
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Susanta K Behura
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| | - Scott Poock
- College of Veterinary Medicine, University of Missouri, Columbia, Missouri, USA
| | - Thomas E Spencer
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
16
|
Zhang Q, Hao J, Li G. Deletion of Prl7d1 causes placental defects at mid-pregnancy in mice. Mol Reprod Dev 2019; 86:696-713. [PMID: 31012985 DOI: 10.1002/mrd.23148] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 03/16/2019] [Accepted: 03/29/2019] [Indexed: 12/29/2022]
Abstract
Prolactin family 7, subfamily d, member 1 (Prl7d1), a member of the expanding prolactin family, is mainly expressed in the placental junctional zone (including trophoblast giant cells and spongiotrophoblast cells) with peak expression observed at 12 days postcoitum (dpc) in mice. Previous studies have shown that PRL7D1 is a key mediator of angiogenesis in vitro; however, its physiological roles in placental development in vivo have not been characterized. To address this issue, we deleted Prl7d1 in mice and demonstrated that its absence results in reduced litter size and fertility. Histologically, Prl7d1 mutants exhibited striking placental abnormalities at 12.5 dpc, including a reduction in the proportion of labyrinth layers and a significant increase in decidual natural killer cells, glycogen trophoblasts, and trophoblast giant cells in the junctional zone. Moreover, placentas from Prl7d1-null mice displayed a thickened decidual spiral artery. Notably, these negative effects were more pronounced in male fetuses. Further RNA-sequencing analysis showed that Prl7d1 deletion results in significant differences in the placental transcriptome profile between the two sexes of fetuses. Together, this study demonstrates that Prl7d1 possesses antiangiogenic properties in deciduas and inhibits the development of junctional zone, which potentially alters the functional capacity of the placenta to support optimal fetal growth. Moreover, of note, the role of Prl7d1 in the placenta is regulated in a fetal sex-specific manner.
Collapse
Affiliation(s)
- Qiong Zhang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jie Hao
- Experimental Research Center, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Gang Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Inoue A, Chen Z, Yin Q, Zhang Y. Maternal Eed knockout causes loss of H3K27me3 imprinting and random X inactivation in the extraembryonic cells. Genes Dev 2018; 32:1525-1536. [PMID: 30463900 PMCID: PMC6295166 DOI: 10.1101/gad.318675.118] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/22/2018] [Indexed: 11/28/2022]
Abstract
In this study, Inoue et al. investigated the regulatory mechanisms and functions of the maternal H3K27me3 mechanism. They found that maternal Eed, an essential component of the Polycomb group complex 2 (PRC2), is required for establishing H3K27me3 imprinting, and their results also reveal unique XCI dynamics in the absence of Xist imprinting. Genomic imprinting is essential for mammalian development. Recent studies have revealed that maternal histone H3 Lys27 trimethylation (H3K27me3) can mediate DNA methylation-independent genomic imprinting. However, the regulatory mechanisms and functions of this new imprinting mechanism are largely unknown. Here we demonstrate that maternal Eed, an essential component of the Polycomb group complex 2 (PRC2), is required for establishing H3K27me3 imprinting. We found that all H3K27me3-imprinted genes, including Xist, lose their imprinted expression in Eed maternal knockout (matKO) embryos, resulting in male-biased lethality. Surprisingly, although maternal X-chromosome inactivation (XmCI) occurs in Eed matKO embryos at preimplantation due to loss of Xist imprinting, it is resolved at peri-implantation. Ultimately, both X chromosomes are reactivated in the embryonic cell lineage prior to random XCI, and only a single X chromosome undergoes random XCI in the extraembryonic cell lineage. Thus, our study not only demonstrates an essential role of Eed in H3K27me3 imprinting establishment but also reveals a unique XCI dynamic in the absence of Xist imprinting.
Collapse
Affiliation(s)
- Azusa Inoue
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Zhiyuan Chen
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Qiangzong Yin
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Harvard Stem Cell Institute, Boston, Massachusetts 02115, USA
| |
Collapse
|
18
|
Abstract
Fundamental differences exist between males and females, encompassing anatomy, physiology, behaviour, and genetics. Such differences undoubtedly play a part in the well documented, yet poorly understood, disparity in disease susceptibility between the sexes. Although traditionally attributed to gonadal sex hormone effects, recent work has begun to shed more light on the contribution of genetics - and in particular the sex chromosomes - to these sexual dimorphisms. Here, we explore the accumulating evidence for a significant genetic component to mammalian sexual dimorphism through the paradigm of sex chromosome evolution. The differences between the extant X and Y chromosomes, at both a sequence and regulatory level, arose across 166 million years. A functional result of these differences is cell autonomous sexual dimorphism. By understanding the process that changed a pair of homologous ancestral autosomes into the extant mammalian X and Y, we believe it easier to consider the mechanisms that may contribute to hormone-independent male-female differences. We highlight key roles for genes with homologues present on both sex chromosomes, where the X-linked copy escapes X chromosome inactivation. Finally, we summarise current experimental paradigms and suggest areas for developments to further increase our understanding of cell autonomous sexual dimorphism in the context of health and disease.
Collapse
Affiliation(s)
- Daniel M Snell
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - James M A Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
19
|
Xu Y, Luo X, Fang Z, Zheng X, Zeng Y, Zhu C, Gu J, Tang F, Hu Y, Hu G, Jin Y, Li H. Transcription coactivator Cited1 acts as an inducer of trophoblast-like state from mouse embryonic stem cells through the activation of BMP signaling. Cell Death Dis 2018; 9:924. [PMID: 30206204 PMCID: PMC6134011 DOI: 10.1038/s41419-018-0991-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 07/16/2018] [Accepted: 08/01/2018] [Indexed: 12/31/2022]
Abstract
Trophoblast lineages, precursors of the placenta, are essential for post-implantation embryo survival. However, the regulatory network of trophoblast development remains incompletely understood. Here, we report that Cited1, a transcription coactivator, is a robust inducer for trophoblast-like state from mouse embryonic stem cells (ESCs). Depletion of Cited1 in ESCs compromises the trophoblast lineage specification induced by BMP signaling. In contrast, overexpression of Cited1 in ESCs induces a trophoblast-like state with elevated expression of trophoblast marker genes in vitro and generation of trophoblastic tumors in vivo. Furthermore, global transcriptome profile analysis indicates that ectopic Cited1 activates a trophoblast-like transcriptional program in ESCs. Mechanistically, Cited1 interacts with Bmpr2 and Smad4 to activate the Cited1–Bmpr2–Smad1/5/8 axis in the cytoplasm and Cited1–Smad4–p300 complexes in the nucleus, respectively. Collectively, our results show that Cited1 plays an important role in regulating trophoblast lineage specification through activating the BMP signaling pathway.
Collapse
Affiliation(s)
- Yanli Xu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Xinlong Luo
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China.,KU Leuven Department of Development and Regeneration, Stem Cell Institute Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Zhuoqing Fang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, 200032, Shanghai, China
| | - Xiaofeng Zheng
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Yanwu Zeng
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Chaonan Zhu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Junjie Gu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Fan Tang
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Yanqin Hu
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China
| | - Guang Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
| | - Ying Jin
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China. .,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, CAS Center for Excellence in Molecular Cell Science, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yueyang Road, 200032, Shanghai, China.
| | - Hui Li
- Basic Clinical Research Center, Renji Hospital, Department of Histology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai JiaoTong University School of Medicine, 225 South Chongqing Road, 200025, Shanghai, China.
| |
Collapse
|
20
|
Woods L, Perez-Garcia V, Hemberger M. Regulation of Placental Development and Its Impact on Fetal Growth-New Insights From Mouse Models. Front Endocrinol (Lausanne) 2018; 9:570. [PMID: 30319550 PMCID: PMC6170611 DOI: 10.3389/fendo.2018.00570] [Citation(s) in RCA: 275] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 09/06/2018] [Indexed: 01/01/2023] Open
Abstract
The placenta is the chief regulator of nutrient supply to the growing embryo during gestation. As such, adequate placental function is instrumental for developmental progression throughout intrauterine development. One of the most common complications during pregnancy is insufficient growth of the fetus, a problem termed intrauterine growth restriction (IUGR) that is most frequently rooted in a malfunctional placenta. Together with conventional gene targeting approaches, recent advances in screening mouse mutants for placental defects, combined with the ability to rapidly induce mutations in vitro and in vivo by CRISPR-Cas9 technology, has provided new insights into the contribution of the genome to normal placental development. Most importantly, these data have demonstrated that far more genes are required for normal placentation than previously appreciated. Here, we provide a summary of common types of placental defects in established mouse mutants, which will help us gain a better understanding of the genes impacting on human placentation. Based on a recent mouse mutant screen, we then provide examples on how these data can be mined to identify novel molecular hubs that may be critical for placental development. Given the close association between placental defects and abnormal cardiovascular and brain development, these functional nodes may also shed light onto the etiology of birth defects that co-occur with placental malformations. Taken together, recent insights into the regulation of mouse placental development have opened up new avenues for research that will promote the study of human pregnancy conditions, notably those based on defects in placentation that underlie the most common pregnancy pathologies such as IUGR and pre-eclampsia.
Collapse
Affiliation(s)
- Laura Woods
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Vicente Perez-Garcia
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Vicente Perez-Garcia
| | - Myriam Hemberger
- Epigenetics Programme, The Babraham Institute, Cambridge, United Kingdom
- Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
- Myriam Hemberger
| |
Collapse
|
21
|
Abstract
OBJECTIVES The aim of this study was to identify an epithelial cell line isolated from the spontaneous differentiation of totipotent pig epiblast cells. METHODS PICM-31 and its colony-cloned derivative cell line, PICM-31A, were established from the culture and differentiation of an epiblast mass isolated from an 8-day-old pig blastocyst. The cell lines were analyzed by transmission electron microscopy, marker gene expression, and mass spectroscopy-based proteomics. RESULTS The PICM-31 cell lines were continuously cultured and could be successively colony cloned. They spontaneously self-organized into acinarlike structures. Transmission electron microscopy indicated that the cell lines' cells were epithelial and filled with secretory granules. Candidate gene expression analysis of the cells showed an exocrine pancreatic profile that included digestive enzyme expression, for example, carboxypeptidase A1, and expression of the fetal marker, α-fetoprotein. Pancreatic progenitor marker expression included pancreatic and duodenal homeobox 1, NK6 homeobox 1, and pancreas-specific transcription factor 1a, but not neurogenin 3. Proteomic analysis of cellular proteins confirmed the cells' production of digestive enzymes and showed that the cells expressed cytokeratins 8 and 18. CONCLUSIONS The PICM-31 cell lines provide in vitro models of fetal pig pancreatic exocrine cells. They are the first demonstration of continuous cultures, that is, cell lines, of nontransformed pig pancreas cells.
Collapse
|
22
|
Del Valle I, Buonocore F, Duncan AJ, Lin L, Barenco M, Parnaik R, Shah S, Hubank M, Gerrelli D, Achermann JC. A genomic atlas of human adrenal and gonad development. Wellcome Open Res 2017. [PMID: 28459107 DOI: 10.12688/wellcomeopenres.11253.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND In humans, the adrenal glands and gonads undergo distinct biological events between 6-10 weeks post conception (wpc), such as testis determination, the onset of steroidogenesis and primordial germ cell development. However, relatively little is currently known about the genetic mechanisms underlying these processes. We therefore aimed to generate a detailed genomic atlas of adrenal and gonad development across these critical stages of human embryonic and fetal development. METHODS RNA was extracted from 53 tissue samples between 6-10 wpc (adrenal, testis, ovary and control). Affymetrix array analysis was performed and differential gene expression was analysed using Bioconductor. A mathematical model was constructed to investigate time-series changes across the dataset. Pathway analysis was performed using ClueGo and cellular localisation of novel factors confirmed using immunohistochemistry. RESULTS Using this approach, we have identified novel components of adrenal development (e.g. ASB4, NPR3) and confirmed the role of SRY as the main human testis-determining gene. By mathematical modelling time-series data we have found new genes up-regulated with SOX9 in the testis (e.g. CITED1), which may represent components of the testis development pathway. We have shown that testicular steroidogenesis has a distinct onset at around 8 wpc and identified potential novel components in adrenal and testicular steroidogenesis (e.g. MGARP, FOXO4, MAP3K15, GRAMD1B, RMND2), as well as testis biomarkers (e.g. SCUBE1). We have also shown that the developing human ovary expresses distinct subsets of genes (e.g. OR10G9, OR4D5), but enrichment for established biological pathways is limited. CONCLUSION This genomic atlas is revealing important novel aspects of human development and new candidate genes for adrenal and reproductive disorders.
Collapse
Affiliation(s)
- Ignacio Del Valle
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Federica Buonocore
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Andrew J Duncan
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Lin Lin
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Martino Barenco
- Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rahul Parnaik
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sonia Shah
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia.,Institute of Cardiovascular Science, University College London, London, UK
| | - Mike Hubank
- The Centre for Molecular Pathology, Royal Marsden Hospital, Sutton, UK
| | - Dianne Gerrelli
- Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child Health, London, UK
| | - John C Achermann
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
23
|
Del Valle I, Buonocore F, Duncan AJ, Lin L, Barenco M, Parnaik R, Shah S, Hubank M, Gerrelli D, Achermann JC. A genomic atlas of human adrenal and gonad development. Wellcome Open Res 2017; 2:25. [PMID: 28459107 PMCID: PMC5407452 DOI: 10.12688/wellcomeopenres.11253.2] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Background: In humans, the adrenal glands and gonads undergo distinct biological events between 6-10 weeks post conception (wpc), such as testis determination, the onset of steroidogenesis and primordial germ cell development. However, relatively little is currently known about the genetic mechanisms underlying these processes. We therefore aimed to generate a detailed genomic atlas of adrenal and gonad development across these critical stages of human embryonic and fetal development. Methods: RNA was extracted from 53 tissue samples between 6-10 wpc (adrenal, testis, ovary and control). Affymetrix array analysis was performed and differential gene expression was analysed using Bioconductor. A mathematical model was constructed to investigate time-series changes across the dataset. Pathway analysis was performed using ClueGo and cellular localisation of novel factors confirmed using immunohistochemistry. Results: Using this approach, we have identified novel components of adrenal development (e.g.
ASB4,
NPR3) and confirmed the role of
SRY as the main human testis-determining gene. By mathematical modelling time-series data we have found new genes up-regulated with
SOX9 in the testis (e.g.
CITED1), which may represent components of the testis development pathway. We have shown that testicular steroidogenesis has a distinct onset at around 8 wpc and identified potential novel components in adrenal and testicular steroidogenesis (e.g.
MGARP,
FOXO4,
MAP3K15,
GRAMD1B,
RMND2), as well as testis biomarkers (e.g.
SCUBE1). We have also shown that the developing human ovary expresses distinct subsets of genes (e.g.
OR10G9,
OR4D5), but enrichment for established biological pathways is limited. Conclusion: This genomic atlas is revealing important novel aspects of human development and new candidate genes for adrenal and reproductive disorders.
Collapse
Affiliation(s)
- Ignacio Del Valle
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Federica Buonocore
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Andrew J Duncan
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Lin Lin
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Martino Barenco
- Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Rahul Parnaik
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sonia Shah
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia.,Institute of Cardiovascular Science, University College London, London, UK
| | - Mike Hubank
- The Centre for Molecular Pathology, Royal Marsden Hospital, Sutton, UK
| | - Dianne Gerrelli
- Developmental Biology and Cancer, UCL Great Ormond Street Institute of Child Health, London, UK
| | - John C Achermann
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, London, UK
| |
Collapse
|
24
|
Sun X, Han Q, Luo H, Pan X, Ji Y, Yang Y, Chen H, Wang F, Lai W, Guan X, Zhang Q, Tang Y, Chu J, Yu J, Shou W, Deng Y, Li X. Profiling analysis of long non-coding RNAs in early postnatal mouse hearts. Sci Rep 2017; 7:43485. [PMID: 28266538 PMCID: PMC5339910 DOI: 10.1038/srep43485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/24/2017] [Indexed: 01/02/2023] Open
Abstract
Mammalian cardiomyocytes undergo a critical hyperplastic-to-hypertrophic growth transition at early postnatal age, which is important in establishing normal physiological function of postnatal hearts. In the current study, we intended to explore the role of long non-coding (lnc) RNAs in this transitional stage. We analyzed lncRNA expression profiles in mouse hearts at postnatal day (P) 1, P7 and P28 via microarray. We identified 1,146 differentially expressed lncRNAs with more than 2.0-fold change when compared the expression profiles of P1 to P7, P1 to P28, and P7 to P28. The neighboring genes of these differentially expressed lncRNAs were mainly involved in DNA replication-associated biological processes. We were particularly interested in one novel cardiac-enriched lncRNA, ENSMUST00000117266, whose expression was dramatically down-regulated from P1 to P28 and was also sensitive to hypoxia, paraquat, and myocardial infarction. Knockdown ENSMUST00000117266 led to a significant increase of neonatal mouse cardiomyocytes in G0/G1 phase and reduction in G2/M phase, suggesting that ENSMUST00000117266 is involved in regulating cardiomyocyte proliferative activity and is likely associated with hyperplastic-to-hypertrophic growth transition. In conclusion, our data have identified a large group of lncRNAs presented in the early postnatal mouse heart. Some of these lncRNAs may have important functions in cardiac hyperplastic-to-hypertrophic growth transition.
Collapse
Affiliation(s)
- Xiongshan Sun
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qi Han
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hongqin Luo
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaodong Pan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yan Ji
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yao Yang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hanying Chen
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fangjie Wang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wenjing Lai
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiao Guan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qi Zhang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yuan Tang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jianhong Chu
- Suzhou Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Weinian Shou
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China.,Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
25
|
Kwon J, Kim NH, Choi I. ROCK activity regulates functional tight junction assembly during blastocyst formation in porcine parthenogenetic embryos. PeerJ 2016; 4:e1914. [PMID: 27077008 PMCID: PMC4830244 DOI: 10.7717/peerj.1914] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 03/18/2016] [Indexed: 11/20/2022] Open
Abstract
The Rho-associated coiled-coil-containing protein serine/threonine kinases 1 and 2 (ROCK1 and ROCK2) are Rho subfamily GTPase downstream effectors that regulate cell migration, intercellular adhesion, cell polarity, and cell proliferation by stimulating actin cytoskeleton reorganization. Inhibition of ROCK proteins affects specification of the trophectoderm (TE) and inner cell mass (ICM) lineages, compaction, and blastocyst cavitation. However, the molecules involved in blastocyst formation are not known. Here, we examined developmental competence and levels of adherens/tight junction (AJ/TJ) constituent proteins, such as CXADR, OCLN, TJP1, and CDH1, as well as expression of their respective mRNAs, after treating porcine parthenogenetic four-cell embryos with Y-27632, a specific inhibitor of ROCK, at concentrations of 0, 10, 20, 100 µM for 24 h. Following this treatment, the blastocyst development rates were 39.1, 20.7, 10.0, and 0% respectively. In embryos treated with 20 µM treatment, expression levels of CXADR, OCLN, TJP1, and CDH1 mRNA and protein molecules were significantly reduced (P < 0.05). FITC-dextran uptake assay revealed that the treatment caused an increase in TE TJ permeability. Interestingly, the majority of the four-cell and morula embryos treated with 20 µM Y-27643 for 24 h showed defective compaction and cavitation. Taken together, our results indicate that ROCK activity may differentially affect assembly of AJ/TJs as well as regulate expression of genes encoding junctional proteins.
Collapse
Affiliation(s)
- Jeongwoo Kwon
- Department of Animal Science, Chungbuk National University , Cheongju Chungbuk , South Korea
| | - Nam-Hyung Kim
- Department of Animal Science, Chungbuk National University , Cheongju Chungbuk , South Korea
| | - Inchul Choi
- Department of Animal and Dairy Sciences, Chungnam National University , Daejeon Chungnam , South Korea
| |
Collapse
|
26
|
Li S, Moore AK, Zhu J, Li X, Zhou H, Lin J, He Y, Xing F, Pan Y, Bohler HC, Ding J, Cooney AJ, Lan Z, Lei Z. Ggnbp2 Is Essential for Pregnancy Success via Regulation of Mouse Trophoblast Stem Cell Proliferation and Differentiation. Biol Reprod 2016; 94:41. [PMID: 26764350 PMCID: PMC4787627 DOI: 10.1095/biolreprod.115.136358] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 11/16/2015] [Accepted: 01/07/2016] [Indexed: 01/16/2023] Open
Abstract
The Ggnbp2 null mutant embryos died in utero between Embryonic Days 13.5 to 15.5 with dysmorphic placentae, characterized by excessive nonvascular cell nests consisting of proliferative trophoblastic tissue and abundant trophoblast stem cells (TSCs) in the labyrinth. Lethality of Ggnbp2 null embryos was caused by insufficient placental perfusion as a result of remarkable decreases in both fetal and maternal blood vessels in the labyrinth. These defects were accompanied by a significant elevation of c-Met expression and phosphorylation and its downstream effector Stat3 activation. Knockdown of Ggnbp2 in wild-type TSCs in vitro provoked the proliferation but delayed the differentiation with an upregulation of c-Met expression and an enhanced phosphorylation of c-Met and Stat3. In contrast, overexpression of Ggnbp2 in wild-type TSCs exhibited completely opposite effects compared to knockdown TSCs. These results suggest that loss of GGNBP2 in the placenta aberrantly overactivates c-Met-Stat3 signaling, alters TSC proliferation and differentiation, and ultimately compromises the structure of placental vascular labyrinth. Our studies for the first time demonstrate that GGNBP2 is an essential factor for pregnancy success acting through the maintenance of a balance of TSC proliferation and differentiation during placental development.
Collapse
Affiliation(s)
- Shengqiang Li
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Andrew K Moore
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Jia Zhu
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Xian Li
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Huaxin Zhou
- Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, University of Louisville School of Dentistry, Louisville, Kentucky
| | - Jing Lin
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Yan He
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Fengying Xing
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Yangbin Pan
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Henry C Bohler
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| | - Jixiang Ding
- Birth Defects Center, Department of Molecular, Cellular and Craniofacial Biology, University of Louisville School of Dentistry, Louisville, Kentucky
| | - Austin J Cooney
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas
| | - Zijian Lan
- Division of Life Sciences and Center for Nutrigenomics & Applied Animal Nutrition, Alltech Inc., Nicholasville, Kentucky
| | - Zhenmin Lei
- Department of OB/GYN & Women's Health, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
27
|
Kulinski TM, Casari MRT, Guenzl PM, Wenzel D, Andergassen D, Hladik A, Datlinger P, Farlik M, Theussl HC, Penninger JM, Knapp S, Bock C, Barlow DP, Hudson QJ. Imprinted expression in cystic embryoid bodies shows an embryonic and not an extra-embryonic pattern. Dev Biol 2015; 402:291-305. [PMID: 25912690 PMCID: PMC4454777 DOI: 10.1016/j.ydbio.2015.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 04/08/2015] [Accepted: 04/16/2015] [Indexed: 12/24/2022]
Abstract
A large subset of mammalian imprinted genes show extra-embryonic lineage (EXEL) specific imprinted expression that is restricted to placental trophectoderm lineages and to visceral yolk sac endoderm (ysE). Isolated ysE provides a homogenous in vivo model of a mid-gestation extra-embryonic tissue to examine the mechanism of EXEL-specific imprinted gene silencing, but an in vitro model of ysE to facilitate more rapid and cost-effective experiments is not available. Reports indicate that ES cells differentiated into cystic embryoid bodies (EBs) contain ysE, so here we investigate if cystic EBs model ysE imprinted expression. The imprinted expression pattern of cystic EBs is shown to resemble fetal liver and not ysE. To investigate the reason for this we characterized the methylome and transcriptome of cystic EBs in comparison to fetal liver and ysE, by whole genome bisulphite sequencing and RNA-seq. Cystic EBs show a fetal liver pattern of global hypermethylation and low expression of repeats, while ysE shows global hypomethylation and high expression of IAPEz retroviral repeats, as reported for placenta. Transcriptome analysis confirmed that cystic EBs are more similar to fetal liver than ysE and express markers of early embryonic endoderm. Genome-wide analysis shows that ysE shares epigenetic and repeat expression features with placenta. Contrary to previous reports, we show that cystic EBs do not contain ysE, but are more similar to the embryonic endoderm of fetal liver. This explains why cystic EBs reproduce the imprinted expression seen in the embryo but not that seen in the ysE.
Collapse
Affiliation(s)
- Tomasz M Kulinski
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| | - M Rita T Casari
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| | - Philipp M Guenzl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| | - Daniel Wenzel
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3, 1030 Vienna, Austria.
| | - Daniel Andergassen
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| | - Anastasiya Hladik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria; Department of Medicine 1, Laboratory of Infection Biology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Paul Datlinger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| | - H-Christian Theussl
- IMP/IMBA Transgenic Service, Institute of Molecular Pathology (IMP), Dr. Bohr Gasse 7, 1030 Vienna, Austria.
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Dr. Bohr Gasse 3, 1030 Vienna, Austria.
| | - Sylvia Knapp
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria; Department of Medicine 1, Laboratory of Infection Biology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| | - Denise P Barlow
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| | - Quanah J Hudson
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH-BT25.3, 1090 Vienna, Austria.
| |
Collapse
|
28
|
Beketaev I, Zhang Y, Kim EY, Yu W, Qian L, Wang J. Critical role of YY1 in cardiac morphogenesis. Dev Dyn 2015; 244:669-80. [PMID: 25703143 DOI: 10.1002/dvdy.24263] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/13/2015] [Accepted: 02/16/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Yin Yang 1 (YY1), the only DNA binding polycomb group protein, was reported to regulate cardiomyocyte differentiation during early cardiac mesoderm development. However, whether it contributes to cardiac morphogenesis at later developmental stage(s) during embryogenesis is unknown. RESULTS We excised YY1 in murine hearts during embryogenesis using two temporal-spatially controlled cre activation approaches, and revealed critical roles of YY1 in cardiac structural formation. Alpha-myosin heavy chain-cre (α-MHC-cre)-mediated cardiomyocyte-specific ablation of YY1 (MHC-YY1) resulted in perinatal death of mutant mice, and Nkx2.5-cre-mediated YY1 null embryos (Nkx2.5-YY1) died embryonically. In the Nkx2.5-YY1 mutants, the endocardial cushions (ECs) of both atrioventricular canal (AVC) and outflow tract (OFT) were hypoplastic due to decreased proliferation and increased apoptosis. Endothelial-to-mesenchymal transition (EMT) progress was also compromised in ECs. Nkx2.5-YY1 mutant hearts had normal formation of extracellular matrix, suggesting that the impaired EMT resulted from the direct loss of YY1. We further uncovered that a number of factors that are involved in normal cardiogenesis were downstream targets of YY1. CONCLUSIONS YY1 plays a critical role in cardiac development and occupies a high-level position within the hierarchy of the cardiac transcriptional network that governs normal cardiogenesis.
Collapse
Affiliation(s)
- Ilimbek Beketaev
- Center for Stem Cell Engineering, Department of Basic Research Laboratories, Texas Heart Institute at St. Luke's Episcopal Hospital, Houston, Texas
| | | | | | | | | | | |
Collapse
|
29
|
Abnormal development of placenta in HtrA1-deficient mice. Dev Biol 2015; 397:89-102. [DOI: 10.1016/j.ydbio.2014.10.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 10/14/2014] [Accepted: 10/18/2014] [Indexed: 01/31/2023]
|
30
|
Depoix CL, Debiève F, Hubinont C. Inhibin alpha gene expression in human trophoblasts is regulated by interactions between TFAP2 and cAMP signaling pathways. Mol Reprod Dev 2014; 81:1009-18. [PMID: 25358080 DOI: 10.1002/mrd.22421] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 08/27/2014] [Indexed: 01/09/2023]
Abstract
Inhibin α (Inha) gene expression is regulated, in rat granulosa cells, via a cyclic 3',5'-adenosine monophosphate (AMP)-response element (CRE) found in a region of the promoter that is homologous to the human INHA promoter. We previously found that during in vitro cytotrophoblast differentiation, human INHA gene expression was regulated by TFAP2A via association with an AP-2 site located upstream of this CRE. The aim of this study was to evaluate if the human INHA gene was also regulated by cAMP in trophoblasts, and to investigate the possible crosstalk between TFAP2 and cAMP signaling pathways in the regulation of INHA gene expression. Treatment with cAMP or forskolin increased INHA mRNA expression by 7- and 2-fold in primary cytotrophoblasts and choriocarcinoma-derived BeWo cells, respectively. Treatment with the protein kinase A inhibitor H-89 reduced forskolin-induced luciferase activity by ∼40% in BeWo cells transfected with an INHA promoter-driven luciferase reporter vector. TFAP2 overexpression increased basal luciferase activity, whereas the dominant repressor KCREB abolished it. Surprisingly, mutation of the CRE also eliminated the TFAP2-induced transcription, although TFAP2 overexpression was still able to increase forskolin-induced luciferase activity when the AP-2 binding site, but not the CRE site, was mutated. Thus, INHA gene expression is upregulated by cAMP via CRE in human trophoblasts, and TFAP2 regulates this expression by interacting with CRE.
Collapse
Affiliation(s)
- Christophe L Depoix
- Département d'obstétrique, Institut de recherche expérimentale et clinique (IREC), Université Catholique de Louvain, Brussels, Belgium
| | | | | |
Collapse
|
31
|
Knott JG, Paul S. Transcriptional regulators of the trophoblast lineage in mammals with hemochorial placentation. Reproduction 2014; 148:R121-36. [PMID: 25190503 DOI: 10.1530/rep-14-0072] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mammalian reproduction is critically dependent on the trophoblast cell lineage, which assures proper establishment of maternal-fetal interactions during pregnancy. Specification of trophoblast cell lineage begins with the development of the trophectoderm (TE) in preimplantation embryos. Subsequently, other trophoblast cell types arise with the progression of pregnancy. Studies with transgenic animal models as well as trophoblast stem/progenitor cells have implicated distinct transcriptional and epigenetic regulators in trophoblast lineage development. This review focuses on our current understanding of transcriptional and epigenetic mechanisms regulating specification, determination, maintenance and differentiation of trophoblast cells.
Collapse
Affiliation(s)
- Jason G Knott
- Developmental Epigenetics LaboratoryDepartment of Animal Science, Michigan State University, East Lansing, Michigan 48824, USADepartment of Pathology and Laboratory MedicineInstitute of Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | - Soumen Paul
- Developmental Epigenetics LaboratoryDepartment of Animal Science, Michigan State University, East Lansing, Michigan 48824, USADepartment of Pathology and Laboratory MedicineInstitute of Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, Kansas 66160, USA
| |
Collapse
|
32
|
Cited2 is required in trophoblasts for correct placental capillary patterning. Dev Biol 2014; 392:62-79. [DOI: 10.1016/j.ydbio.2014.04.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 04/21/2014] [Accepted: 04/23/2014] [Indexed: 01/14/2023]
|
33
|
Nolte MJ, Wang Y, Deng JM, Swinton PG, Wei C, Guindani M, Schwartz RJ, Behringer RR. Functional analysis of limb transcriptional enhancers in the mouse. Evol Dev 2014; 16:207-23. [PMID: 24920384 DOI: 10.1111/ede.12084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Transcriptional enhancers are genomic sequences bound by transcription factors that act together with basal transcriptional machinery to regulate gene transcription. Several high-throughput methods have generated large datasets of tissue-specific enhancer sequences with putative roles in developmental processes. However, few enhancers have been deleted from the genome to determine their roles in development. To understand the roles of two enhancers active in the mouse embryonic limb bud we deleted them from the genome. Although the genes regulated by these enhancers are unknown, they were selected because they were identified in a screen for putative limb bud-specific enhancers associated with p300, an acetyltransferase that participates in protein complexes that promote active transcription, and because the orthologous human enhancers (H1442 and H280) drive distinct lacZ expression patterns in limb buds of embryonic day (E) 11.5 transgenic mice. We show that the orthologous mouse sequences, M1442 and M280, regulate dynamic expression in the developing limb. Although significant transcriptional differences in enhancer-proximal genes in embryonic limb buds accompany the deletion of M1442 and M280 no gross limb malformations during embryonic development were observed, demonstrating that M1442 and M280 are not required for mouse limb development. However, M280 is required for the development and/or maintenance of body size; M280 mice are significantly smaller than controls. M280 also harbors an "ultraconserved" sequence that is identical between human, rat, and mouse. This is the first report of a phenotype resulting from the deletion of an ultraconserved element. These studies highlight the importance of determining enhancer regulatory function by experiments that manipulate them in situ and suggest that some of an enhancer's regulatory capacities may be developmentally tolerated rather than developmentally required.
Collapse
Affiliation(s)
- Mark J Nolte
- Graduate Program in Genes and Development, University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA; Department of Genetics, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Hirasawa R, Matoba S, Inoue K, Ogura A. Somatic donor cell type correlates with embryonic, but not extra-embryonic, gene expression in postimplantation cloned embryos. PLoS One 2013; 8:e76422. [PMID: 24146866 PMCID: PMC3797840 DOI: 10.1371/journal.pone.0076422] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 08/23/2013] [Indexed: 11/19/2022] Open
Abstract
The great majority of embryos generated by somatic cell nuclear transfer (SCNT) display defined abnormal phenotypes after implantation, such as an increased likelihood of death and abnormal placentation. To gain better insight into the underlying mechanisms, we analyzed genome-wide gene expression profiles of day 6.5 postimplantation mouse embryos cloned from three different cell types (cumulus cells, neonatal Sertoli cells and fibroblasts). The embryos retrieved from the uteri were separated into embryonic (epiblast) and extraembryonic (extraembryonic ectoderm and ectoplacental cone) tissues and were subjected to gene microarray analysis. Genotype- and sex-matched embryos produced by in vitro fertilization were used as controls. Principal component analysis revealed that whereas the gene expression patterns in the embryonic tissues varied according to the donor cell type, those in extraembryonic tissues were relatively consistent across all groups. Within each group, the embryonic tissues had more differentially expressed genes (DEGs) (>2-fold vs. controls) than did the extraembryonic tissues (P<1.0×10–26). In the embryonic tissues, one of the common abnormalities was upregulation of Dlk1, a paternally imprinted gene. This might be a potential cause of the occasional placenta-only conceptuses seen in SCNT-generated mouse embryos (1–5% per embryos transferred in our laboratory), because dysregulation of the same gene is known to cause developmental failure of embryos derived from induced pluripotent stem cells. There were also some DEGs in the extraembryonic tissues, which might explain the poor development of SCNT-derived placentas at early stages. These findings suggest that SCNT affects the embryonic and extraembryonic development differentially and might cause further deterioration in the embryonic lineage in a donor cell-specific manner. This could explain donor cell-dependent variations in cloning efficiency using SCNT.
Collapse
Affiliation(s)
| | - Shogo Matoba
- RIKEN BioResouce Center, Tsukuba, Ibaraki, Japan
| | - Kimiko Inoue
- RIKEN BioResouce Center, Tsukuba, Ibaraki, Japan
- Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Atsuo Ogura
- RIKEN BioResouce Center, Tsukuba, Ibaraki, Japan
- Graduate School of Life and Environmental Science, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
35
|
Méniel V, Song F, Phesse T, Young M, Poetz O, Parry L, Jenkins JR, Williams GT, Dunwoodie SL, Watson A, Clarke AR. Cited1 deficiency suppresses intestinal tumorigenesis. PLoS Genet 2013; 9:e1003638. [PMID: 23935526 PMCID: PMC3731217 DOI: 10.1371/journal.pgen.1003638] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 06/01/2013] [Indexed: 11/23/2022] Open
Abstract
Conditional deletion of Apc in the murine intestine alters crypt-villus architecture and function. This process is accompanied by multiple changes in gene expression, including upregulation of Cited1, whose role in colorectal carcinogenesis is unknown. Here we explore the relevance of Cited1 to intestinal tumorigenesis. We crossed Cited1 null mice with Apc(Min/+) and AhCre(+)Apc(fl/fl) mice and determined the impact of Cited1 deficiency on tumour growth/initiation including tumour multiplicity, cell proliferation, apoptosis and the transcriptome. We show that Cited1 is up-regulated in both human and murine tumours, and that constitutive deficiency of Cited1 increases survival in Apc(Min/+) mice from 230.5 to 515 days. However, paradoxically, Cited1 deficiency accentuated nearly all aspects of the immediate phenotype 4 days after conditional deletion of Apc, including an increase in cell death and enhanced perturbation of differentiation, including of the stem cell compartment. Transcriptome analysis revealed multiple pathway changes, including p53, PI3K and Wnt. The activation of Wnt through Cited1 deficiency correlated with increased transcription of β-catenin and increased levels of dephosphorylated β-catenin. Hence, immediately following deletion of Apc, Cited1 normally restrains the Wnt pathway at the level of β-catenin. Thus deficiency of Cited1 leads to hyper-activation of Wnt signaling and an exaggerated Wnt phenotype including elevated cell death. Cited1 deficiency decreases intestinal tumourigenesis in Apc(Min/+) mice and impacts upon a number of oncogenic signaling pathways, including Wnt. This restraint imposed by Cited1 is consistent with a requirement for Cited1 to constrain Wnt activity to a level commensurate with optimal adenoma formation and maintenance, and provides one mechanism for tumour repression in the absence of Cited1.
Collapse
Affiliation(s)
- Valérie Méniel
- School of Biological Sciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Fei Song
- Department of Gastroenterology, Institute of Translational Medicine, The Henry Wellcome Laboratory, University of Liverpool, England, United Kingdom
- Institute of Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Toby Phesse
- Cell Signaling and Cell Death, Walter and Eliza Hall Institute for Medical Research, Melbourne, Victoria, Australia
| | - Madeleine Young
- School of Biological Sciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - Oliver Poetz
- Natural and Medical Sciences Institute at the University of Tuebingen, Reutlingen, Germany
| | - Lee Parry
- School of Biological Sciences, Cardiff University, Cardiff, Wales, United Kingdom
| | - John R. Jenkins
- Department of Gastroenterology, Institute of Translational Medicine, The Henry Wellcome Laboratory, University of Liverpool, England, United Kingdom
| | - Geraint T. Williams
- School of Medicine, Cardiff University, Heath Park, Cardiff, Wales, United Kingdom
| | - Sally L. Dunwoodie
- Developmental and Stem Cell Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Kensington, Sydney, New South Wales, Australia
| | - Alastair Watson
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, United Kingdom
| | - Alan R. Clarke
- School of Biological Sciences, Cardiff University, Cardiff, Wales, United Kingdom
| |
Collapse
|
36
|
Epigenetic regulation of placental endocrine lineages and complications of pregnancy. Biochem Soc Trans 2013; 41:701-9. [DOI: 10.1042/bst20130002] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A defining feature of mammals is the development in utero of the fetus supported by the constant flow of nutrients from the mother obtained via a specialized organ: the placenta. The placenta is also a major endocrine organ that synthesizes vast quantities of hormones and cytokines to instruct both maternal and fetal physiology. Nearly 20 years ago, David Haig and colleagues proposed that placental hormones were likely targets of the epigenetic process of genomic imprinting in response to the genetic conflicts imposed by in utero development [Haig (1993) Q. Rev. Biol. 68, 495–532]. There are two simple mechanisms through which genomic imprinting could regulate placental hormones. First, imprints could directly switch on or off alleles of specific genes. Secondly, imprinted genes could alter the expression of placental hormones by regulating the development of placental endocrine lineages. In mice, the placental hormones are synthesized in the trophoblast giant cells and spongiotrophoblast cells of the mature placenta. In the present article, I review the functional role of imprinted genes in regulating these endocrine lineages, which lends support to Haig's original hypothesis. I also discuss how imprinting defects in the placenta may adversely affect the health of the fetus and its mother during pregnancy and beyond.
Collapse
|
37
|
Sferruzzi-Perri AN, Vaughan OR, Forhead AJ, Fowden AL. Hormonal and nutritional drivers of intrauterine growth. Curr Opin Clin Nutr Metab Care 2013; 16:298-309. [PMID: 23340010 DOI: 10.1097/mco.0b013e32835e3643] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE OF REVIEW Size at birth is critical in determining life expectancy with both small and large neonates at risk of shortened life spans. This review examines the hormonal and nutritional drivers of intrauterine growth with emphasis on the role of foetal hormones as nutritional signals in utero. RECENT FINDINGS Nutrients drive intrauterine growth by providing substrate for tissue accretion, whereas hormones regulate nutrient distribution between foetal oxidative metabolism and mass accumulation. The main hormonal drivers of intrauterine growth are insulin, insulin-like growth factors and thyroid hormones. Together with leptin and cortisol, these hormones control cellular nutrient uptake and the balance between accretion and differentiation in regulating tissue growth. They also act indirectly via the placenta to alter the materno-foetal supply of nutrients and oxygen. By responding to nutrient and oxygen availability, foetal hormones optimize the survival and growth of the foetus with respect to its genetic potential, particularly during adverse conditions. However, changes in the intrauterine growth of individual tissues may alter their function permanently. SUMMARY In both normal and compromised pregnancies, intrauterine growth is determined by multiple hormonal and nutritional drivers which interact to produce a specific pattern of intrauterine development with potential lifelong consequences for health.
Collapse
Affiliation(s)
- Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK.
| | | | | | | |
Collapse
|
38
|
Tunster SJ, Jensen AB, John RM. Imprinted genes in mouse placental development and the regulation of fetal energy stores. Reproduction 2013; 145:R117-37. [PMID: 23445556 DOI: 10.1530/rep-12-0511] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Imprinted genes, which are preferentially expressed from one or other parental chromosome as a consequence of epigenetic events in the germline, are known to functionally converge on biological processes that enable in utero development in mammals. Over 100 imprinted genes have been identified in the mouse, the majority of which are both expressed and imprinted in the placenta. The purpose of this review is to provide a summary of the current knowledge regarding imprinted gene function in the mouse placenta. Few imprinted genes have been assessed with respect to their dosage-related action in the placenta. Nonetheless, current data indicate that imprinted genes converge on two key functions of the placenta, nutrient transport and placental signalling. Murine studies may provide a greater understanding of certain human pathologies, including low birth weight and the programming of metabolic diseases in the adult, and complications of pregnancy, such as pre-eclampsia and gestational diabetes, resulting from fetuses carrying abnormal imprints.
Collapse
Affiliation(s)
- S J Tunster
- Cardiff School of Biosciences, Cardiff University, Museum Avenue, Cardiff, Wales CF10 3AX, UK
| | | | | |
Collapse
|
39
|
Vijaya M, Manikandan J, Parakalan R, Dheen ST, Kumar SD, Tay SSW. Differential gene expression profiles during embryonic heart development in diabetic mice pregnancy. Gene 2012; 516:218-27. [PMID: 23287646 DOI: 10.1016/j.gene.2012.12.071] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 12/09/2012] [Indexed: 11/19/2022]
Abstract
Congenital heart defects (CHD) are one of the most common defects in offspring of diabetic mothers. There is a clear association between maternal diabetes and CHD; however the underlying molecular mechanism remains unknown. We hypothesized that maternal diabetes affects with the expression of early developmental genes that regulate the essential developmental processes of the heart, thereby resulting in the pathogenesis of CHD. We analyzed genome-wide expression profiling in the developing heart of embryos from diabetic and control mice by using the oligonucleotide microarray. Microarray analysis revealed that a total of 878 genes exhibited more than 1.5 fold changes in expression level in the hearts of experimental embryos in either E13.5 or E15.5 compared with their respective controls. Expression pattern of genes that is differentially expressed in the developing heart was further examined by the real-time reverse transcriptase-polymerase chain reaction. Several genes involved in a number of molecular signaling pathways such as apoptosis, proliferation, migration and differentiation in the developing heart were differentially expressed in embryos of diabetic pregnancy. It is concluded that altered expression of several genes involved in heart development may contribute to CHD in offspring of diabetic mothers.
Collapse
Affiliation(s)
- Murugaiyan Vijaya
- Department of Anatomy, National University of Singapore, Singapore 117597, Singapore
| | | | | | | | | | | |
Collapse
|
40
|
A placenta for life. Reprod Biomed Online 2012; 25:5-11. [PMID: 22578825 DOI: 10.1016/j.rbmo.2012.03.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 11/22/2022]
Abstract
The chorioallantoic placenta is the defining organ of eutherians that has enabled prolonged intrauterine gestation. As such, normal placental development and function are essential for mammalian reproductive success. Reflecting the key role of this organ in providing nutrients to the embryo, the characteristic cell type that forms substantial parts of the placenta is called 'trophoblast' (from Greek trephein 'to feed' and blastos 'germinator'). However, in addition to regulating nutrient supply, the placenta also exerts a number of other pivotal functions that highlight the importance of normal trophoblast differentiation for a successful pregnancy. In this guest symposium, 'Trophoblast Development', several contributors summarize insights gained from recent studies in the mouse that have advanced our understanding of trophoblast biology. This includes how the earliest trophoblast cells are set aside to expand in a stem- or progenitor-cell compartment under tight genetic and epigenetic control and how subsequent differentiation into the various placental cell types is controlled to ensure normal placentation. The relevance of these contributions range from early developmental cell fate decisions, stem cell biology and placental development for healthy pregnancy to the impact of placental failures on long-term health, with important clinical implications for assisted reproductive technology procedures and pregnancy-associated complications.
Collapse
|
41
|
Banerjee I, Zhang J, Moore-Morris T, Lange S, Shen T, Dalton ND, Gu Y, Peterson KL, Evans SM, Chen J. Thymosin beta 4 is dispensable for murine cardiac development and function. Circ Res 2011; 110:456-64. [PMID: 22158707 DOI: 10.1161/circresaha.111.258616] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Thymosin beta 4 (Tβ4) is a 43-amino acid factor encoded by an X-linked gene. Recent studies have suggested that Tβ4 is a key factor in cardiac development, growth, disease, epicardial integrity, and blood vessel formation. Cardiac-specific short hairpin (sh)RNA knockdown of tβ4 has been reported to result in embryonic lethality at E14.5-16.5, with severe cardiac and angiogenic defects. However, this shRNA tβ4-knockdown model did not completely abrogate Tβ4 expression. To completely ablate Tβ4 and to rule out the possibility of off-target effects associated with shRNA gene silencing, further studies of global or cardiac-specific knockouts are critical. OBJECTIVE We examined the role of Tβ4 in developing and adult heart through global and cardiac specific tβ4-knockout mouse models. METHODS AND RESULTS Global tβ4-knockout mice were born at mendelian ratios and exhibited normal heart and blood vessel formation. Furthermore, in adult global tβ4-knockout mice, cardiac function, capillary density, expression of key cardiac fetal and angiogenic genes, epicardial marker expression, and extracellular matrix deposition were indistinguishable from that of controls. Tissue-specific tβ4-deficient mice, generated by crossing tβ4-floxed mice to Nkx2.5-Cre and αMHC-Cre, were also found to have no phenotype. CONCLUSIONS We conclude that Tβ4 is dispensable for embryonic viability, heart development, coronary vessel development, and adult myocardial function.
Collapse
Affiliation(s)
- Indroneal Banerjee
- Department of Medicine, University of California-San Diego, La Jolla, 92093, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Denda K, Nakao-Wakabayashi K, Okamoto N, Kitamura N, Ryu JY, Tagawa YI, Ichisaka T, Yamanaka S, Komada M. Nrk, an X-linked protein kinase in the germinal center kinase family, is required for placental development and fetoplacental induction of labor. J Biol Chem 2011; 286:28802-28810. [PMID: 21715335 DOI: 10.1074/jbc.m111.258160] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The complete mechanism of labor induction in eutherian mammals remains unclear. Although important roles for the fetus and placenta in triggering labor have been proposed, no gene has been shown to be required in the fetus/placenta for labor induction. Here we show that Nrk, an X-linked gene encoding a Ser/Thr kinase of the germinal center kinase family, is essential in the fetus/placenta for labor in mice. Nrk was specifically expressed in the spongiotrophoblast layer, a fetus-derived region of the placenta, and Nrk disruption caused dysregulated overgrowth of the layer. Due to preferential inactivation of the paternally derived X chromosome in placenta, Nrk heterozygous mutant placentas exhibited a similar defect to that in Nrk-null tissues when the wild-type allele was paternally derived. However, the phenotype was weaker than in Nrk-null placentas due to leaky Nrk expression from the inactivated X chromosome. Crossing of Nrk-null females to wild-type and Nrk-null males, as well as uterine transfer of Nrk-null fetuses to wild-type females, revealed that pregnant mice exhibit a severe defect in delivery when all fetuses/placentas are Nrk-null. In addition, Nrk was not expressed in female reproductive tissues such as the uterus and ovary, as well as the fetal amnion and yolk sac, in pregnant mice. Progesterone and estrogen levels in the maternal circulation and placenta, which control the timing of labor, were unaffected upon Nrk disruption. We thus provide evidence for a novel labor-inducing fetoplacental signal that depends on the X chromosome and possibly arises from the placenta.
Collapse
Affiliation(s)
- Kimitoshi Denda
- Department of Biological Sciences, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | | | - Naoki Okamoto
- Department of Biological Sciences, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Naomi Kitamura
- Department of Biological Sciences, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Je-Young Ryu
- Biomolecular Engineering, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Yoh-Ichi Tagawa
- Biomolecular Engineering, Tokyo Institute of Technology, Yokohama 226-8501, Japan
| | - Tomoko Ichisaka
- Department of Reprogramming Science, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Shinya Yamanaka
- Department of Reprogramming Science, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Masayuki Komada
- Department of Biological Sciences, Tokyo Institute of Technology, Yokohama 226-8501, Japan.
| |
Collapse
|
43
|
Novitskaya T, Baserga M, de Caestecker MP. Organ-specific defects in insulin-like growth factor and insulin receptor signaling in late gestational asymmetric intrauterine growth restriction in Cited1 mutant mice. Endocrinology 2011; 152:2503-16. [PMID: 21486933 PMCID: PMC3100618 DOI: 10.1210/en.2010-1385] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Late gestational placental insufficiency resulting in asymmetric intrauterine organ growth restriction (IUGR) is associated with an increased incidence of diabetes, cardiovascular and renal disease in adults. The molecular mechanisms mediating these defects are poorly understood. To explore this, we investigated the mechanisms leading to IUGR in Cited1 knockout mice, a genetic model of late gestational placental insufficiency. We show that loss of placental Cited1 leads to asymmetric IUGR with decreased liver, lung, and kidney sizes and preservation of fetal brain weight. IGF and insulin signaling regulate embryonic organ growth. IGF-I and IGF-II protein and mRNA expression are reduced in livers, lungs, and kidneys of embryonic d 18.5 embryos with IUGR. Decreased IGF-I is associated with reduced activating phosphorylation of the type 1 IGF receptor (pIGF-IR) in the kidney, whereas reduced IGF-II is associated with decreased phosphorylation of the insulin receptor (pIR) in the lung. In contrast, decreased pIR is associated with reduced IGF-I but not IGF-II in the liver. However, pancreatic β-cell mass and serum insulin levels are also decreased in mice with IUGR, suggesting that hepatic IR signaling may be regulated by alterations in fetal insulin production. These findings contrast with observations in IUGR fetal brains in which there is no change in IGF-IR/IR phosphorylation, and IGF-I and IGF-II expression is actually increased. In conclusion, IUGR disrupts normal fetal IGF and insulin production and is associated with organ-specific defects in IGF-IR and IR signaling that may regulate asymmetric IUGR in late gestational placental insufficiency.
Collapse
Affiliation(s)
- Tatiana Novitskaya
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, Tennessee 37232, USA
| | | | | |
Collapse
|
44
|
Mammalian Llgl2 is necessary for proper branching morphogenesis during placental development. Mol Cell Biol 2011; 31:2920-33. [PMID: 21606200 DOI: 10.1128/mcb.05431-11] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cell polarity plays a critical role in the development of all metazoans; however, the mechanisms of cell polarity and the specific role of cell polarity pathways in mammalian organisms are still poorly understood. Lethal giant larvae (Lgl) is an apical-basal polarity gene identified in Drosophila, where it functions as a tumor suppressor controlling self-renewal and differentiation of progenitor cells. There are two orthologs of Lgl in mammalian genomes: Llgl1 and Llgl2. While mammalian Lgls are assumed to be tumor suppressor genes, little is known about their function in vivo. Here we report the functional analysis of murine Llgl2. We generated Llgl2(-/-) mice and found that Llgl2 functions as a polarity protein required for proper branching morphogenesis during placental development. Llgl2(-/-) pups are born as runts but quickly catch up in size and grow into normal-size adults. Surprisingly, no prominent phenotypes or spontaneous tumors were observed in adult Llgl2(-/-) mice. Analyses of placental trophoblasts reveal a critical role for Llgl2 in cell polarization and polarized cell invasion. We conclude that mammalian Llgl2 is required for proper polarized invasion of trophoblasts and efficient branching morphogenesis during placental development, but, unlike its Drosophila ortholog, it does not function as a canonical tumor suppressor gene.
Collapse
|
45
|
Lopes Floro K, Artap ST, Preis JI, Fatkin D, Chapman G, Furtado MB, Harvey RP, Hamada H, Sparrow DB, Dunwoodie SL. Loss of Cited2 causes congenital heart disease by perturbing left–right patterning of the body axis. Hum Mol Genet 2010; 20:1097-110. [DOI: 10.1093/hmg/ddq554] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
46
|
Degrelle SA, Lê Cao KA, Heyman Y, Everts RE, Campion E, Richard C, Ducroix-Crépy C, Tian XC, Lewin HA, Renard JP, Robert-Granié C, Hue I. A small set of extra-embryonic genes defines a new landmark for bovine embryo staging. Reproduction 2010; 141:79-89. [PMID: 20926692 DOI: 10.1530/rep-10-0174] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Axis specification in mouse is determined by a sequence of reciprocal interactions between embryonic and extra-embryonic tissues so that a few extra-embryonic genes appear as 'patterning' the embryo. Considering these interactions as essential, but lacking in most mammals the genetically driven approaches used in mouse and the corresponding patterning mutants, we examined whether a molecular signature originating from extra-embryonic tissues could relate to the developmental stage of the embryo proper and predict it. To this end, we have profiled bovine extra-embryonic tissues at peri-implantation stages, when gastrulation and early neurulation occur, and analysed the subsequent expression profiles through the use of predictive methods as previously reported for tumour classification. A set of six genes (CALM1, CPA3, CITED1, DLD, HNRNPDL, and TGFB3), half of which had not been previously associated with any extra-embryonic feature, appeared significantly discriminative and mainly dependent on embryonic tissues for its faithful expression. The predictive value of this set of genes for gastrulation and early neurulation stages, as assessed on naive samples, was remarkably high (93%). In silico connected to the bovine orthologues of the mouse patterning genes, this gene set is proposed as a new trait for embryo staging. As such, this will allow saving the bovine embryo proper for molecular or cellular studies. To us, it offers as well new perspectives for developmental phenotyping and modelling of embryonic/extra-embryonic co-differentiation.
Collapse
Affiliation(s)
- Séverine A Degrelle
- INRA-ENVA, UMR 1198 Biologie du Développement et Reproduction, Domaine de Vilvert, Jouy en Josas, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nadra K, Quignodon L, Sardella C, Joye E, Mucciolo A, Chrast R, Desvergne B. PPARgamma in placental angiogenesis. Endocrinology 2010; 151:4969-81. [PMID: 20810566 DOI: 10.1210/en.2010-0131] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a nuclear receptor involved in diverse biological processes including adipocyte differentiation, glucose homeostasis, and inflammatory responses. Analyses of PPARγ knockout animals have been so far preempted by the early embryonic death of PPARγ-/- embryos as a consequence of the severe alteration of their placental vasculature. Using Sox2Cre/PPARγL2/L2 mice, we obtained fully viable PPARγ-null mice through specific and total epiblastic gene deletion, thereby demonstrating that the placental defect is the unique cause of PPARγ-/- embryonic lethality. The vasculature defects observed in PPARγ-/- placentas at embryonic d 9.5 correlated with an unsettled balance of pro- and antiangiogenic factors as demonstrated by increased levels of proliferin (Prl2c2, PLF) and decreased levels of proliferin-related protein (Prl7d1, PRP), respectively. To analyze the role of PPARγ in the later stage of placental development, when its expression peaks, we treated pregnant wild-type mice with the PPARγ agonist rosiglitazone. This treatment resulted in a disorganization of the placental layers and an altered placental microvasculature, accompanied by the decreased expression of proangiogenic genes such as Prl2c2, vascular endothelial growth factor, and Pecam1. Together our data demonstrate that PPARγ plays a pivotal role in controlling placental vascular proliferation and contributes to its termination in late pregnancy.
Collapse
Affiliation(s)
- Karim Nadra
- Center for Integrative Genomics, Department of Medical Genetics and Electron Microscopy Platform, University of Lausanne, CH-1015 Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
48
|
Fauque P, Mondon F, Letourneur F, Ripoche MA, Journot L, Barbaux S, Dandolo L, Patrat C, Wolf JP, Jouannet P, Jammes H, Vaiman D. In vitro fertilization and embryo culture strongly impact the placental transcriptome in the mouse model. PLoS One 2010; 5:e9218. [PMID: 20169163 PMCID: PMC2821408 DOI: 10.1371/journal.pone.0009218] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2009] [Accepted: 01/24/2010] [Indexed: 12/17/2022] Open
Abstract
Background Assisted Reproductive Technologies (ART) are increasingly used in humans; however, their impact is now questioned. At blastocyst stage, the trophectoderm is directly in contact with an artificial medium environment, which can impact placental development. This study was designed to carry out an in-depth analysis of the placental transcriptome after ART in mice. Methodology/Principal Findings Blastocysts were transferred either (1) after in vivo fertilization and development (control group) or (2) after in vitro fertilization and embryo culture. Placentas were then analyzed at E10.5. Six percent of transcripts were altered at the two-fold threshold in placentas of manipulated embryos, 2/3 of transcripts being down-regulated. Strikingly, the X-chromosome harbors 11% of altered genes, 2/3 being induced. Imprinted genes were modified similarly to the X. Promoter composition analysis indicates that FOXA transcription factors may be involved in the transcriptional deregulations. Conclusions For the first time, our study shows that in vitro fertilization associated with embryo culture strongly modify the placental expression profile, long after embryo manipulations, meaning that the stress of artificial environment is memorized after implantation. Expression of X and imprinted genes is also greatly modulated probably to adapt to adverse conditions. Our results highlight the importance of studying human placentas from ART.
Collapse
Affiliation(s)
- Patricia Fauque
- Service d'Histologie-Embryologie, Biologie de la Reproduction, Hôpital Cochin, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Sriraman V, Sinha M, Richards JS. Progesterone receptor-induced gene expression in primary mouse granulosa cell cultures. Biol Reprod 2009; 82:402-12. [PMID: 19726735 DOI: 10.1095/biolreprod.109.077610] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The progesterone receptor (PGR) is induced by luteinizing hormone (LH) in granulosa cells of preovulatory follicles, and the PGR-A isoform is essential for ovulation based on the phenotypes of Pgr isoform-specific knockout mice. Although several genes regulated by PGR-A in vivo have been identified, whether these genes are primary targets of PGR-A or if their expression also depends on other signaling molecules that are induced by the LH surge has not been resolved. Therefore, to identify genes that are either induced or repressed by PGR in the absence of LH-mediated signaling cascades, we infected primary cultures of mouse granulosa cells with either PGR-A or PGR-B adenoviral vectors without or with R-5020 as a PGR ligand. Total RNA was extracted from infected cells at 16 h and analyzed by Affymetrix Mouse 430 2.0 microarrays. PGR-A in the presence or absence of ligand significantly induced approximately 50 genes 2-fold or more (local pooled error test at P <or= 0.01). Fewer and different genes were induced by PGR-B in the absence of ligand. Edn1, Apoa1, and Cited1 were primarily regulated by PGR-A as verified by additional RT-PCR analyses, suppression by the PGR antagonist RU486, and the lack of induction by protein kinase A, protein kinase C, or epidermal growth factor (EGF)-like factors pathways. PGR regulation of these genes was confirmed further by gene expression analyses in hormonally primed Pgr mutant mouse ovaries. Because Edn1, Apoa1, and Cited1 are known to regulate angiogenesis, PGR may affect the neovascularization of follicles that is initiated with ovulation.
Collapse
|
50
|
Comparative systems biology of human and mouse as a tool to guide the modeling of human placental pathology. Mol Syst Biol 2009; 5:279. [PMID: 19536202 PMCID: PMC2710868 DOI: 10.1038/msb.2009.37] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2009] [Accepted: 05/13/2009] [Indexed: 11/29/2022] Open
Abstract
Placental abnormalities are associated with two of the most common and serious complications of human pregnancy, maternal preeclampsia (PE) and fetal intrauterine growth restriction (IUGR), each disorder affecting ∼5% of all pregnancies. An important question for the use of the mouse as a model for studying human disease is the degree of functional conservation of genetic control pathways from human to mouse. The human and mouse placenta show structural similarities, but there have been no systematic attempts to assess their molecular similarities or differences. We collected protein and mRNA expression data through shot-gun proteomics and microarray expression analysis of the highly vascular exchange region, microdissected from the human and mouse near-term placenta. Over 7000 ortholog genes were detected with 70% co-expressed in both species. Close to 90% agreement was found between our human proteomic results and 1649 genes assayed by immunohistochemistry for expression in the human placenta in the Human Protein Atlas. Interestingly, over 80% of genes known to cause placental phenotypes in mouse are co-expressed in human. Several of these phenotype-associated proteins form a tight protein–protein interaction network involving 15 known and 34 novel candidate proteins also likely important in placental structure and/or function. The entire data are available as a web-accessible database to guide the informed development of mouse models to study human disease.
Collapse
|