1
|
Pirali A, Jafarpour F, Hajian M, Hosseini Moghaddam SH, Moradi R, Tanhaie-Vash N, Rahimi Andani M, Izadi T, Shiralian-Esfahani H, Safaeinejad Z, Kues W, Nasr-Esfahani MH, Eghbalsaied S. Editing the CYP19 Gene in Goat Embryos Using CRISPR/Cas9 and Somatic Cell Nuclear Transfer Techniques. Cell Reprogram 2025; 27:86-93. [PMID: 40126138 DOI: 10.1089/cell.2024.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
The Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR/Cas9) system is revolutionizing genome engineering and is expected to bring significant advancements in livestock traits, including the treatment of genetic diseases. This study focuses on CRISPR/Cas9-mediated modifications of the CYP19 gene, which encodes aromatase, an enzyme crucial for converting testosterone to estrogen and essential for steroid metabolism. Guide RNAs (gRNAs) were designed to target the CYP19 gene and cloned into the pX459 vector. The recombinant plasmid was then electrotransfected into fibroblast cells from a Lori-Bakhtiari buck, and these transfected cells were used for embryo production via somatic cell nuclear transfer (SCNT). The cloned embryos were evaluated for their progression through embryonic stages, showing no significant difference in blastocyst development between knock-out and unedited groups. The knockout efficiency was 78.4% in cells and 68.9% in goat blastocysts, demonstrating the successful depletion of CYP19. We successfully achieved a high rate of CYP19 gene-edited embryos through the combined application of cell electrotransfection and SCNT technologies, while maintaining the normal developmental rate of the embryos. These embryos can be used for transfer to generate knock-out goats, providing a foundation for further studies on CYP19's role in male fertility and production traits.
Collapse
Affiliation(s)
- Ahmad Pirali
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Faculty of Agricultural Sciences, Department of Animal Science, University of Guilan, Rasht, Iran
| | - Farnoosh Jafarpour
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mehdi Hajian
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Reza Moradi
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Nima Tanhaie-Vash
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohsen Rahimi Andani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Tayebeh Izadi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hanieh Shiralian-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Zahra Safaeinejad
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Wilfried Kues
- Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Neustadt Rbge, Germany
| | - Mohammad-Hossein Nasr-Esfahani
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Shahin Eghbalsaied
- Department of Animal Biotechnology, Reproductive Biomedicine Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
- Department of Animal Science, Isfahan (Khorasgan) Branch, Islamic Azad University, Tehran, Iran
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
2
|
Vacher CM, Tsompanidis A, Firestein MR, Penn AA. Neuroactive steroid exposure impacts neurodevelopment: Comparison of human and rodent placental contribution. J Neuroendocrinol 2025:e13489. [PMID: 39789736 DOI: 10.1111/jne.13489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
The placenta is a fetal endocrine organ that secretes many neuroactive factors, including steroids, that play critical roles in brain development. The study of the placenta-brain axis and the links between placental function and brain development represents an emerging research area dubbed "neuroplacentology." The placenta drives many circulating fetal steroids to very high levels during gestation. Recent studies have highlighted the critical role of placental steroids in shaping specific brain structures and behaviors. This review uses a cross-species framework to discuss the genomic factors, in-utero environmental changes, and placental conditions that alter placental steroidogenesis, leading to changes in early developmental trajectories relevant for psychiatric conditions such as autism, in a sex-linked manner.
Collapse
Affiliation(s)
- Claire-Marie Vacher
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Morgan R Firestein
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, USA
| | - Anna A Penn
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
- New York Presbyterian Morgan Stanley Children's Hospital, New York, New York, USA
| |
Collapse
|
3
|
Nunes LGA, Rosario FJ, Urschitz J. In vivo placental gene modulation via sonoporation. Placenta 2024:S0143-4004(24)00688-X. [PMID: 39477696 PMCID: PMC12014858 DOI: 10.1016/j.placenta.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/25/2024] [Accepted: 10/21/2024] [Indexed: 11/29/2024]
Abstract
Placental dysregulation frequently results in pregnancy complications that impact fetal well-being and potentially predispose the infant to diseases later in life. Thus, efforts to understand the molecular mechanisms underlying placental disorders are crucial to aid the development of effective treatments to restore placental function. Currently, the most common methods used for trophoblast-specific gene modulation in the laboratory are transgenic animals and lentiviral trophectoderm transduction. The generation of transgenic animal lines is costly and requires a considerable amount of time to generate and maintain, while the integration preference of lentiviruses, actively transcribed genes, may result in genotoxicity. Therefore, there is much interest in the development of non-viral in vivo transfection techniques for use in both research and clinical settings. Herein, we describe a non-viral, minimally invasive method for in vivo placental gene modulation through sonoporation, an ultrasound-mediated transfection technique wherein the application of ultrasound on target tissues is used to direct the uptake of DNA vectors. In this method, plasmids are bound to lipid microbubbles, which are then injected into the maternal bloodstream and ultimately delivered to the placenta when subjected to low-frequency ultrasound. Syncytiotrophoblasts are directly exposed to maternal blood and, therefore highly accessible to therapeutic agents in the maternal circulation. This technique can be used to modulate gene expression and, subsequently, the function of the placenta, circumventing the requirement to generate transgenic animals. Sonoporation also offers a safer alternative to existing viral techniques, making it not only an advantageous research tool but also a potentially adaptable technique in clinical settings.
Collapse
Affiliation(s)
- Lance G A Nunes
- Institute for Biogenesis Research, University of Hawai'i, Honolulu, HI, United States
| | - Fredrick J Rosario
- Department of Obstetrics and Gynecology, University of Colorado, Anschutz Medical Campus, Aurora, CO, United States
| | - Johann Urschitz
- Institute for Biogenesis Research, University of Hawai'i, Honolulu, HI, United States.
| |
Collapse
|
4
|
Guengerich FP, Tateishi Y, McCarty KD, Yoshimoto FK. Updates on Mechanisms of Cytochrome P450 Catalysis of Complex Steroid Oxidations. Int J Mol Sci 2024; 25:9020. [PMID: 39201706 PMCID: PMC11354347 DOI: 10.3390/ijms25169020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/14/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
Cytochrome P450 (P450) enzymes dominate steroid metabolism. In general, the simple C-hydroxylation reactions are mechanistically straightforward and are generally agreed to involve a perferryl oxygen species (formally FeO3+). Several of the steroid transformations are more complex and involve C-C bond scission. We initiated mechanistic studies with several of these (i.e., 11A1, 17A1, 19A1, and 51A1) and have now established that the dominant modes of catalysis for P450s 19A1 and 51A1 involve a ferric peroxide anion (i.e., Fe3+O2¯) instead of a perferryl ion complex (FeO3+), as demonstrated with 18O incorporation studies. P450 17A1 is less clear. The indicated P450 reactions all involve sequential oxidations, and we have explored the processivity of these multi-step reactions. P450 19A1 is distributive, i.e., intermediate products dissociate and reassociate, but P450s 11A1 and 51A1 are highly processive. P450 17A1 shows intermediate processivity, as expected from the release of 17-hydroxysteroids for the biosynthesis of key molecules, and P450 19A1 is very distributive. P450 11B2 catalyzes a processive multi-step oxidation process with the complexity of a chemical closure of an intermediate to a locked lactol form.
Collapse
Affiliation(s)
- F. Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (Y.T.); (K.D.M.)
| | - Yasuhiro Tateishi
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (Y.T.); (K.D.M.)
| | - Kevin D. McCarty
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; (Y.T.); (K.D.M.)
| | - Francis K. Yoshimoto
- Department of Chemistry, University of Texas at San Antonio, San Antonio, TX 78249, USA;
| |
Collapse
|
5
|
Paixão RV, Silva GF, Caetano AR, Cintra LC, Varela ES, O'Sullivan FLA. Phylogenomic and expression analysis of Colossoma macropomum cyp19a1a and cyp19a1b and their non-classical role in tambaqui sex differentiation. Gene 2022; 843:146795. [PMID: 35961435 DOI: 10.1016/j.gene.2022.146795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/15/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022]
Abstract
The genes coding for Cytochrome P450 aromatase (cyp19a1a and cyp19a1b) and estrogen (E2) receptors (esr1, esr2a and esr2b) play a conserved role in ovarian differentiation and development among teleosts. Classically, the "gonad form" of aromatase, coded by the cyp19a1a, is responsible for the ovarian differentiation in genetic females via ligation and activation of the Esr, which mediates the endocrine and exocrine signaling to allow or block the establishment of the feminine phenotype. However, in neotropical species, studies on the molecular and endocrine processes involved in gonad differentiation as well as on the effects of sex modulators are recent and scarce. In this study, we combined in silico analysis, real-time quantitative PCR (qPCR) assay and quantification of E2 plasma levels of differentiating tambaqui (Colossoma macropomum) to unveil the roles of the paralogs cypa19a1a and cyp19a1b during sex differentiation. Although the synteny of each gene is very conserved among characids, the genomic environment displays striking differences in comparison to model teleost species, with many rearrangements in cyp19a1a and cyp19a1b adjacencies and transposable element traces in both regulatory regions. The high dissimilarity (DI) of SF-1 binding motifs in cyp19a1a (DI = 10.06 to 14.90 %) and cyp19a1b (DI = 8.41 to 13.50 %) regulatory region, respectively, may reflect in an alternative pathway in tambaqui. Indeed, while low transcription of cyp19a1a was detected prior to sex differentiation, the expression of cyp19a1b and esr2a presented a large variation at this phase, which could be associated with sex-specific differential expression. Histological analysis revealed that anti-estradiol treatments did not affect gonadal sex ratios, although Fadrozole (50 mg kg-1 of food) reduced E2 plasma levels (p < 0,005) as well cyp19a1a transcription; and tamoxifen (200 mg kg-1 of food) down regulated both cyp19a1a and cyp19a1b but did not influence E2 levels. Altogether, our results bring into light new insights about the evolutionary fate of cyp19a1 paralogs in neotropical fish, which may have generated uncommon roles for the gonadal and brain forms of cyp19a1 genes and the unexpected lack of effect of endocrine disruptors on tambaqui sexual differentiation.
Collapse
Affiliation(s)
- R V Paixão
- Universidade Federal do Amazonas (UFAM), Programa de Pós-graduação em Ciência Animal e Recursos Pesqueiros, Avenida Rodrigo Otávio, CEP: 69080-900, 6200 Manaus, AM, Brazil
| | - G F Silva
- Embrapa Amazônia Ocidental, Rodovia AM-010, Km 29, Caixa Postal 319, CEP: 69010-790, Brazil
| | - A R Caetano
- Embrapa Recursos Genéticos e Biotecnologia, Final Av. W/5 Norte, C.P. 02372, CEP 70770-917, Brasília, DF, Brazil
| | - L C Cintra
- Embrapa Agricultura Digital, Avenida André Tosselo, 209, Cidade Universitária, CEP: 13083-886, Campinas, SP, Brazil
| | - E S Varela
- Embrapa Pesca e Aquicultura, Av. NS 10, cruzamento com a Av. LO 18 Sentido Norte Loteamento - Água Fria, Palmas, TO 77008-900, Brazil
| | - F L A O'Sullivan
- Embrapa Amazônia Ocidental, Rodovia AM-010, Km 29, Caixa Postal 319, CEP: 69010-790, Brazil.
| |
Collapse
|
6
|
Hong K, Muralimanoharan S, Kwak YT, Mendelson CR. NRF2 Serves a Critical Role in Regulation of Immune Checkpoint Proteins (ICPs) During Trophoblast Differentiation. Endocrinology 2022; 163:bqac070. [PMID: 35596653 PMCID: PMC9197021 DOI: 10.1210/endocr/bqac070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Indexed: 11/19/2022]
Abstract
Using cultured human trophoblast stem cells (hTSCs), mid-gestation human trophoblasts in primary culture, and gene-targeted mice, we tested the hypothesis that the multinucleated syncytiotrophoblast (SynT) serves a critical role in pregnancy maintenance through production of key immune modulators/checkpoint proteins (ICPs) under control of the O2-regulated transcription factor, NRF2/NFE2L2. These ICPs potentially act at the maternal-fetal interface to protect the hemiallogeneic fetus from rejection by the maternal immune system. Using cultured hTSCs, we observed that several ICPs involved in the induction and maintenance of immune tolerance were markedly upregulated during differentiation of cytotrophoblasts (CytTs) to SynT. These included HMOX1, kynurenine receptor, aryl hydrocarbon receptor, PD-L1, and GDF15. Intriguingly, NRF2, C/EBPβ, and PPARγ were markedly induced when CytTs fused to form SynT in a 20% O2 environment. Notably, when hTSCs were cultured in a hypoxic (2% O2) environment, SynT fusion and the differentiation-associated induction of NRF2, C/EBPβ, aromatase (CYP19A1; SynT differentiation marker), and ICPs were blocked. NRF2 knockdown also prevented induction of aromatase, C/EBPβ and the previously mentioned ICPs. Chromatin immunoprecipitation-quantitative PCR revealed that temporal induction of the ICPs in hTSCs and mid-gestation human trophoblasts cultured in 20% O2 was associated with increased binding of endogenous NRF2 to putative response elements within their promoters. Moreover, placentas of 12.5 days postcoitum mice with a global Nrf2 knockout manifested decreased mRNA expression of C/ebpβ, Pparγ, Hmox1, aryl hydrocarbon receptor, and Nqo1, another direct downstream target of Nrf2, compared with wild-type mice. Collectively, these compelling findings suggest that O2-regulated NRF2 serves as a key regulator of ICP expression during SynT differentiation.
Collapse
Affiliation(s)
- Kyunghee Hong
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9038, USA
| | | | - Youn-Tae Kwak
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9038, USA
| | - Carole R Mendelson
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390-9038, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9032, USA
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390-8511, USA
- North Texas March of Dimes Birth Defects Center, University of Texas Southwestern Medical Center, Dallas, TX 75390-9038, USA
| |
Collapse
|
7
|
Rendic SP, Peter Guengerich F. Human cytochrome P450 enzymes 5-51 as targets of drugs and natural and environmental compounds: mechanisms, induction, and inhibition - toxic effects and benefits. Drug Metab Rev 2019; 50:256-342. [PMID: 30717606 DOI: 10.1080/03602532.2018.1483401] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cytochrome P450 (P450, CYP) enzymes have long been of interest due to their roles in the metabolism of drugs, pesticides, pro-carcinogens, and other xenobiotic chemicals. They have also been of interest due to their very critical roles in the biosynthesis and metabolism of steroids, vitamins, and certain eicosanoids. This review covers the 22 (of the total of 57) human P450s in Families 5-51 and their substrate selectivity. Furthermore, included is information and references regarding inducibility, inhibition, and (in some cases) stimulation by chemicals. We update and discuss important aspects of each of these 22 P450s and questions that remain open.
Collapse
Affiliation(s)
| | - F Peter Guengerich
- b Department of Biochemistry , Vanderbilt University School of Medicine , Nashville , TN , USA
| |
Collapse
|
8
|
Wattez JS, Qiao L, Lee S, Natale DRC, Shao J. The platelet-derived growth factor receptor alpha promoter-directed expression of cre recombinase in mouse placenta. Dev Dyn 2019; 248:363-374. [PMID: 30843624 PMCID: PMC6488356 DOI: 10.1002/dvdy.21] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/09/2019] [Accepted: 02/22/2019] [Indexed: 12/12/2022] Open
Abstract
Background Numerous pathologies of pregnancy originate from placental dysfunction. It is essential to understand the functions of key genes in the placenta in order to discern the etiology of placental pathologies. A paucity of animal models that allow conditional and inducible expression of a target gene in the placenta is a major limitation for studying placental development and function. Results To study the platelet‐derived growth factor receptor alpha (PDGFRα)‐directed and tamoxifen‐induced Cre recombinase expression in the placenta, PDGFRα‐CreER mice were crossed with mT/mG dual‐fluorescent reporter mice. The expression of endogenous membrane‐localized enhanced green fluorescent protein (mEGFP) and/or dTomato in the placenta was examined to identify PDGFRα promoter‐directed Cre expression. Pregnant PDGFRα‐CreER;mT/mG mice were treated with tamoxifen at various gestational ages. Upon tamoxifen treatment, reporter protein mEGFP was observed in the junctional zone (JZ) and chorionic plate (CP). Furthermore, a single dose of tamoxifen was sufficient to induce the recombination. Conclusions PDGFRα‐CreER expression is restricted to the JZ and CP of mouse placentas. PDGFRα‐CreER mice provide a useful tool to conditionally knock out or overexpress a target gene in these regions of the mouse placenta. Inducible PDGFRα‐directed Cre expression trophoblasts cells. A single tamoxifen treatment is sufficient to induce the recombination. Valuable tool to temporary knockout or over‐express a target gene in the placenta. Do not require sophisticated system and suitable for ordinary laboratory setting.
Collapse
Affiliation(s)
| | - Liping Qiao
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | - Samuel Lee
- Department of Pediatrics, University of California San Diego, La Jolla, California
| | | | - Jianhua Shao
- Department of Pediatrics, University of California San Diego, La Jolla, California
| |
Collapse
|
9
|
Elston M, Urschitz J. Transposase-mediated gene modulation in the placenta. Placenta 2017; 59 Suppl 1:S32-S36. [PMID: 28778732 PMCID: PMC5682209 DOI: 10.1016/j.placenta.2017.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 07/12/2017] [Accepted: 07/27/2017] [Indexed: 11/27/2022]
Abstract
The placenta is an organ vital to fetal development as well as the maintenance of a healthy pregnancy and plays a crucial role in developmental programming of the fetus. The mechanisms that link intrauterine milieu, fetal health and disease development later in life are poorly understood. Placenta-specific gene modulation, both by generating transgenic animals as well as by developing methods for in vivo genetic modifications is a growing area of interest as this approach provides the opportunity to investigate the role of particular genes or gene networks in regulating placental function and fetal growth. Furthermore, in vivo placental gene transfer may be adapted to treat humans in the future and could be used as an early intervention strategy for a wide range of pregnancy complications. This review is an overview of transposase-based methods available for both transgenic animal generation and in vivo placental gene modifications with an emphasis on piggyBac-based systems.
Collapse
Affiliation(s)
- Marlee Elston
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, Honolulu, HI 96822, United States
| | - Johann Urschitz
- Department of Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, Honolulu, HI 96822, United States.
| |
Collapse
|
10
|
Szalai G, Xu Y, Romero R, Chaiworapongsa T, Xu Z, Chiang PJ, Ahn H, Sundell B, Plazyo O, Jiang Y, Olive M, Wang B, Jacques SM, Qureshi F, Tarca AL, Erez O, Dong Z, Papp Z, Hassan SS, Hernandez-Andrade E, Than NG. In vivo experiments reveal the good, the bad and the ugly faces of sFlt-1 in pregnancy. PLoS One 2014; 9:e110867. [PMID: 25393290 PMCID: PMC4230935 DOI: 10.1371/journal.pone.0110867] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 09/23/2014] [Indexed: 01/06/2023] Open
Abstract
Objective Soluble fms-like tyrosine kinase (sFlt)-1-e15a, a primate-specific sFlt-1-isoform most abundant in the human placenta in preeclampsia, can induce preeclampsia in mice. This study compared the effects of full-length human (h)sFlt-1-e15a with those of truncated mouse (m)sFlt-1(1-3) used in previous preeclampsia studies on pregnancy outcome and clinical symptoms in preeclampsia. Methods Mice were injected with adenoviruses or fiber-mutant adenoviruses overexpressing hsFlt-1-e15a, msFlt-1(1-3) or control GFP under the CMV or CYP19A1 promoters on gestational day 8 (GD8) and GD11. Placentas and pups were delivered by cesarean section, and dams were monitored postpartum. Blood pressure was telemetrically recorded. Urine samples were collected with cystocentesis and examined for albumin/creatinine ratios. Tissue specimens were evaluated for transgene as well as endogenous mFlt-1 and msFlt-1-i13 expression. H&E-, Jones- and PAS-stained kidney sections were histopathologically examined. Placental GFP expression and aortic ring assays were investigated with confocal microscopy. Results Mean arterial blood pressure (MAP) was elevated before delivery in hsFlt-1-e15a-treated mice compared to controls (GD18: ΔMAP = 7.8 mmHg, p = 0.009), while ΔMAP was 12.8 mmHg (GD18, p = 0.005) in msFlt-1(1-3)-treated mice. Urine albumin/creatinine ratio was higher in hsFlt-1-e15a-treated mice than in controls (GD18, p = 0.04; PPD8, p = 0.03), and msFlt-1(1-3)-treated mice had marked proteinuria postpartum (PPD8, p = 4×10−5). Focal glomerular changes were detected in hsFlt-1-e15a and msFlt-1(1-3)-treated mice. Aortic ring microvessel outgrowth was decreased in hsFlt-1-e15a (p = 0.007) and msFlt-1(1-3)-treated (p = 0.02) mice. Full-length msFlt-1-i13 expression was unique for the placenta. In hsFlt-1-e15a-treated mice, the number of pups (p = 0.046), total weight of living pups (p = 0.04) and maternal weights (p = 0.04) were higher than in controls. These differences were not observed in truncated msFlt-1(1-3)-treated mice. Conclusions Truncated msFlt-1(1-3) simulated the preeclampsia-promoting effects of full-length hsFlt-1. MsFlt-1(1-3) had strong effect on maternal endothelium but not on placentas and embryos. In contrast, hsFlt-1-e15a induced preeclampsia-like symptoms; however, it also increased litter size. In accord with the predominant placental expression of hsFlt-1-e15a and msFlt-1-i13, full-length sFlt-1 may have a role in the regulation of embryonic development. These observations point to the difference in the biological effects of full-length and truncated sFlt-1 and the changes in the effect of full-length sFlt-1 during pregnancy, and may have important implications in the management of preeclampsia.
Collapse
Affiliation(s)
- Gabor Szalai
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Yi Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Roberto Romero
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- * E-mail: (RR); (NGT)
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Zhonghui Xu
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Po Jen Chiang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Hyunyoung Ahn
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Birgitta Sundell
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Olesya Plazyo
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Yang Jiang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Mary Olive
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Bing Wang
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Suzanne M. Jacques
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Faisal Qureshi
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Adi L. Tarca
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Computer Science, Wayne State University, Detroit, MI, United States of America
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Offer Erez
- Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Zhong Dong
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
| | - Zoltan Papp
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
| | - Sonia S. Hassan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Edgar Hernandez-Andrade
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States of America
| | - Nandor Gabor Than
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, and Detroit, MI, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States of America
- Maternity Private Department, Kutvolgyi Clinical Block, Semmelweis University, Budapest, Hungary
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
- * E-mail: (RR); (NGT)
| |
Collapse
|
11
|
Placental trophoblast cell differentiation: Physiological regulation and pathological relevance to preeclampsia. Mol Aspects Med 2013; 34:981-1023. [DOI: 10.1016/j.mam.2012.12.008] [Citation(s) in RCA: 238] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/01/2012] [Accepted: 12/19/2012] [Indexed: 12/11/2022]
|
12
|
Strauss L, Rantakari P, Sjögren K, Salminen A, Lauren E, Kallio J, Damdimopoulou P, Boström M, Boström PJ, Pakarinen P, Zhang F, Kujala P, Ohlsson C, Mäkelä S, Poutanen M. Seminal vesicles and urinary bladder as sites of aromatization of androgens in men, evidenced by a CYP19A1‐driven luciferase reporter mouse and human tissue specimens. FASEB J 2012; 27:1342-50. [DOI: 10.1096/fj.12-219048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Leena Strauss
- Department of PhysiologyUniversity of TurkuTurkuFinland
- Institute of BiomedicineTurku Center for Disease ModelingUniversity of TurkuTurkuFinland
- Laboratory of Electron MicroscopyUniversity of TurkuTurkuFinland
| | - Pia Rantakari
- Department of PhysiologyUniversity of TurkuTurkuFinland
- Institute of BiomedicineTurku Center for Disease ModelingUniversity of TurkuTurkuFinland
| | - Klara Sjögren
- Center for Bone and Arthritis ResearchInstitute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Anu Salminen
- Department of PhysiologyUniversity of TurkuTurkuFinland
| | - Eve Lauren
- Department of PhysiologyUniversity of TurkuTurkuFinland
| | - Jenny Kallio
- Department of PhysiologyUniversity of TurkuTurkuFinland
- Institute of BiomedicineTurku Center for Disease ModelingUniversity of TurkuTurkuFinland
| | - Pauliina Damdimopoulou
- Institute of BiomedicineTurku Center for Disease ModelingUniversity of TurkuTurkuFinland
- Functional Foods ForumUniversity of TurkuTurkuFinland
| | - Minna Boström
- Division of UrologyDepartment of SurgeryTurku University HospitalTurkuFinland
| | - Peter J. Boström
- Division of UrologyDepartment of SurgeryTurku University HospitalTurkuFinland
| | - Pirjo Pakarinen
- Department of PhysiologyUniversity of TurkuTurkuFinland
- Institute of BiomedicineTurku Center for Disease ModelingUniversity of TurkuTurkuFinland
| | - FuPing Zhang
- Department of PhysiologyUniversity of TurkuTurkuFinland
- Institute of BiomedicineTurku Center for Disease ModelingUniversity of TurkuTurkuFinland
| | - Paula Kujala
- Department of PathologyTampere University HospitalTampereFinland
| | - Claes Ohlsson
- Center for Bone and Arthritis ResearchInstitute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Sari Mäkelä
- Institute of BiomedicineTurku Center for Disease ModelingUniversity of TurkuTurkuFinland
- Functional Foods ForumUniversity of TurkuTurkuFinland
| | - Matti Poutanen
- Department of PhysiologyUniversity of TurkuTurkuFinland
- Institute of BiomedicineTurku Center for Disease ModelingUniversity of TurkuTurkuFinland
- Center for Bone and Arthritis ResearchInstitute of Medicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
13
|
Kumar P, Mendelson CR. Estrogen-related receptor gamma (ERRgamma) mediates oxygen-dependent induction of aromatase (CYP19) gene expression during human trophoblast differentiation. Mol Endocrinol 2011; 25:1513-26. [PMID: 21757507 DOI: 10.1210/me.2011-1012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Differentiation of human cytotrophoblasts to syncytiotrophoblast and the associated induction of aromatase/hCYP19 gene expression are dependent upon a critical O(2) tension; however, the underlying molecular mechanisms remain undefined. In this study, we provide compelling evidence that expression of the orphan nuclear receptor, estrogen-related receptor γ (ERRγ), is also O(2) dependent, induced during human syncytiotrophoblast differentiation, and plays an obligatory role in the induction of placenta-specific hCYP19I.1 gene expression. Treatment with the selective ERRγ agonist, DY131, or overexpression of ERRγ, stimulated hCYP19 expression in syncytiotrophoblast. Overexpression of ERRγ prevented effects of hypoxia to repress hCYP19 gene expression in cultured trophoblasts. Conversely, small interfering RNA-mediated knockdown of endogenous ERRγ in primary trophoblasts markedly inhibited hCYP19 expression. Promoter and site-directed mutagenesis studies in transfected placental cells identified a nuclear receptor element within placenta-specific hCYP19 promoter I.1 required for ERRγ-stimulated activity. Recruitment of endogenous ERRγ to the nuclear receptor element region in hCYP19 promoter during trophoblast differentiation, assessed by chromatin immunoprecipitation, was prevented by hypoxia. Deferoxamine-induced hypoxia-inducible factor-1α (HIF-1α) levels decreased ERRγ expression, whereas knockdown of endogenous HIF-1α prevented ERRγ suppression by hypoxia. Chromatin immunoprecipitation analysis of trophoblasts cultured in hypoxia revealed recruitment of HIF-1α to one of two putative hypoxia response elements in the ERRγ promoter, providing in vivo evidence of a direct HIF-1α involvement in ERRγ expression. Collectively, these novel findings identify ERRγ as an O(2)-dependent transcription factor and HIF-1α target gene that serves a critical role in the induction of hCYP19 expression during human trophoblast differentiation.
Collapse
Affiliation(s)
- Premlata Kumar
- Department of Biochemistry, North Texas March of Dimes Birth Defects Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9038, USA
| | | |
Collapse
|
14
|
Stimulation of serotonergic 5-HT2A receptor signaling increases placental aromatase (CYP19) activity and expression in BeWo and JEG-3 human choriocarcinoma cells. Placenta 2011; 32:651-656. [PMID: 21703684 DOI: 10.1016/j.placenta.2011.06.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/03/2011] [Accepted: 06/06/2011] [Indexed: 01/23/2023]
Abstract
It is known that serotonin can influence the production and function of sex hormones, such as estrogens. Estrogens are critical for maintenance of pregnancy and regulate placental and fetal development. The key enzyme controlling estrogens synthesis during pregnancy is placental aromatase (CYP19). To better understand the regulation of placental aromatase, this study determined whether serotonin is involved in the regulation of this enzyme. BeWo and JEG-3 choriocarcinoma cells were used as models of the human placental trophoblast to evaluate the effects of serotonin and selective 5-HT(2A) receptor agonists on CYP19 activity and expression. Serotonin and selective 5-HT(2A) receptor agonists as well as PKC activation increased aromatase activity and expression in BeWo and JEG-3 cells. Dexamethasone, which regulates aromatase expression via JAK/STAT activation in certain tissues, had no effect. Increased CYP19 gene transcription by 5-HT(2A) receptor and PKC stimulation was mediated by activation of the placental I.1 aromatase promoter. This study shows that the serotonergic system modulates placental aromatase expression, which would result in altered estrogens biosynthesis in trophoblast cells. Future detailed studies of serotonin-estrogen interactions in placenta are crucial for an improved understanding of the endo-, para- and autocrine role of serotonin during pregnancy and fetal development.
Collapse
|
15
|
Renaud SJ, Karim Rumi MA, Soares MJ. Review: Genetic manipulation of the rodent placenta. Placenta 2011; 32 Suppl 2:S130-5. [PMID: 21256588 DOI: 10.1016/j.placenta.2010.12.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Revised: 12/10/2010] [Accepted: 12/13/2010] [Indexed: 10/18/2022]
Abstract
The principal role of the placenta is the maintenance of pregnancy and promotion of fetal growth and viability. The use of transgenic rodents has greatly enhanced our understanding of placental development and function. However, embryonic lethality is often a confounding variable in determining whether a genetic modification adversely affected placental development. In these cases, it is beneficial to specifically manipulate the placental genome. The purpose of this review is to summarize available methodologies for specific genetic modification of the rodent placenta. By restricting genetic alterations to the trophoblast lineage, it is possible to gain a deeper understanding of placental development that perhaps will lead to gene-targeted therapies to rescue irregular placentation in transgenic animals or in women at high-risk for placenta-associated pregnancy complications.
Collapse
Affiliation(s)
- S J Renaud
- Institute for Reproductive Health and Regenerative Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | | | |
Collapse
|
16
|
Fürbass R, Tomek W, Vanselow J. Upstream stimulating factors 1 and 2 enhance transcription from the placenta-specific promoter 1.1 of the bovine cyp19 gene. BMC Mol Biol 2010; 11:5. [PMID: 20082704 PMCID: PMC2822775 DOI: 10.1186/1471-2199-11-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2009] [Accepted: 01/18/2010] [Indexed: 11/27/2022] Open
Abstract
Background Placenta-derived oestrogens have an impact on the growth and differentiation of the trophoblast, and are involved in processes initiating and facilitating birth. The enzyme that converts androgens into oestrogens, aromatase cytochrome P450 (P450arom), is encoded by the Cyp19 gene. In the placenta of the cow, expression of Cyp19 relies on promoter 1.1 (P1.1). Our recent studies of P1.1 in vitro and in a human trophoblast cell line (Jeg3) revealed that interactions of placental nuclear protein(s) with the E-box element at position -340 are required for full promoter activity. The aim of this work was to identify and characterise the placental E-box (-340)-binding protein(s) (E-BP) as a step towards understanding how the expression of Cyp19 is regulated in the bovine placenta. Results The significance of the E-box was confirmed in cultured primary bovine trophoblasts. We enriched the E-BP from placental nuclear extracts using DNA-affinity Dynabeads and showed by Western blot analysis and supershift EMSA experiments that the E-BP is composed of the transcription factors upstream stimulating factor (USF) 1 and USF2. Depletion of the USFs by RNAi and expression of a dominant-negative USF mutant, were both associated with a significant decrease in P1.1-dependent reporter gene expression. Furthermore, scatter plot analysis of P1.1 activity vs. USF binding to the E-box revealed a strong positive correlation between the two parameters. Conclusion From these results we conclude that USF1 and USF2 are activators of the bovine placenta-specific promoter P1.1 and thus act in the opposite mode as in the case of the non-orthologous human placenta-specific promoter.
Collapse
Affiliation(s)
- Rainer Fürbass
- Research Unit Molecular Biology, Research Institute for the Biology of Farm Animals (FBN), Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany.
| | | | | |
Collapse
|
17
|
Kumar P, Kamat A, Mendelson CR. Estrogen receptor alpha (ERalpha) mediates stimulatory effects of estrogen on aromatase (CYP19) gene expression in human placenta. Mol Endocrinol 2009; 23:784-93. [PMID: 19299445 DOI: 10.1210/me.2008-0371] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A 246-bp region upstream of placenta-specific exon I.1 of the human aromatase (hCYP19) gene mediates placenta-specific, developmental, and O(2) regulation of expression. In this study, trophoblast differentiation and associated induction of CYP19 expression were prevented when cytotrophoblasts were cultured in phenol red-free medium containing charcoal-stripped serum or with the estrogen receptor (ER) antagonist, ICI 182,780, suggesting a stimulatory role of estrogen/ER. ERalpha protein was expressed in human trophoblasts and increased during syncytiotrophoblast differentiation, whereas ERbeta was undetectable. Mutational analysis revealed that an estrogen response element-like sequence (ERE-LS) at -208 bp is required for inductive effects of estradiol/ERalpha on hCYP19I.1 promoter activity in transfected COS-7 cells. Increased binding of syncytiotrophoblast compared with cytotrophoblast nuclear proteins to the ERE-LS was observed in vitro; however, ERalpha antibodies failed to supershift the complex and in vitro-transcribed/translated ERalpha did not bind. Nonetheless, chromatin immunoprecipitation assays in cultured trophoblasts revealed recruitment of endogenous ERalpha to the -255- to -155-bp region containing the ERE-LS before induction of hCYP19 expression; this was inhibited by ICI 182,780. Chromatin immunoprecipitation also revealed increased acetylated histone H3(K9/14) and decreased methylated histone H3(K9) associated with this region during trophoblast differentiation. These modifications were prevented when trophoblasts were incubated with ICI 182,780, suggesting that ERalpha recruitment to the -255- to -155-bp region promotes histone modifications leading to increased hCYP19 transcription. Thus, during trophoblast differentiation, estrogen/ERalpha exerts a positive feedback role, which promotes permissive histone modifications that are associated with induction of hCYP19 gene transcription.
Collapse
Affiliation(s)
- Premlata Kumar
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, 75390-9038, USA
| | | | | |
Collapse
|
18
|
Jin Y, Lu SY, Fresnoza A, Detillieux KA, Duckworth ML, Cattini PA. Differential placental hormone gene expression during pregnancy in a transgenic mouse containing the human growth hormone/chorionic somatomammotropin locus. Placenta 2009; 30:226-35. [PMID: 19168217 DOI: 10.1016/j.placenta.2008.12.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 12/16/2008] [Accepted: 12/18/2008] [Indexed: 01/01/2023]
Abstract
The human (h) growth hormone/chorionic somatomammotropin (GH/CS) gene locus presents a unique model to gain insight into the molecular mechanisms that have allowed a closely related family of genes to be expressed in two distinct cell lineages/tissues: pituitary somatotrophs and placental syncytiotrophoblasts. However, studies of external factors that regulate gene expression have been somewhat limited by (i) a lack of human cell lines expressing endogenous GH or CS appropriately; and (ii) the fact that the GH/CS locus is unique to primates and thus does not exist in rodents. In the current study, a transgenic (171 h GH/CS-TG) mouse was generated containing the intact hGH/CS gene cluster and hGH locus control region (LCR) in a 171-kilobase DNA fragment. Pituitary and placental-specific expression of hGH/CS RNA was detected at embryonic day (E) 18.5. Immunostaining of hGH was seen in somatotrophs of the anterior pituitary beginning in late gestation. The presence of hCS protein was detected in the placental labyrinth in trophoblasts functionally analogous to the syncytiotrophoblast of the chorionic villi. This pattern of gene expression is consistent with the presence of essential components of the hGH/CS LCR. Transcript levels for hCS-A, hCS-B and placental hGH-variant increased in 171 hGH/CS-TG placenta during gestation (E11.5-E18.5), as previously observed in human placental development. Throughout gestation, hCS-A RNA levels were proportionately higher, accounting for 91% of total CS RNA by E18.5, comparable to term human placenta. Finally, the previous correlation between the transcription factor AP-2alpha and hCS RNA expression observed in developing primary human cytotrophoblast cultures, was extended to pregnancy in the 171 hGH/CS-TG mouse. The 171 hGH/CS-TG mouse thus provides a model to investigate hGH/CS gene expression, including in pregnancy.
Collapse
Affiliation(s)
- Y Jin
- Department of Physiology, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB, Canada R3E 0J9
| | | | | | | | | | | |
Collapse
|
19
|
Affiliation(s)
- Saara M. Rawn
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, and the Graduate Program in Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; ,
| | - James C. Cross
- Department of Comparative Biology & Experimental Medicine, Faculty of Veterinary Medicine, and the Graduate Program in Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta T2N 4N1, Canada; ,
| |
Collapse
|
20
|
Abstract
During human gestation, the placental syncytiotrophoblast develops the capacity to synthesize large amounts of estrogen from C(19)-steroids secreted by the fetal adrenals. The conversion of C(19)-steroids to estrogens is catalyzed by aromatase P450 (P450arom), product of the CYP19 gene. The placenta-specific promoter of the hCYP19 gene lies approximately 100,000 bp upstream of the translation initiation site in exon II. In studies using transgenic mice and transfected human trophoblast cells we have defined a 246-bp region upstream of placenta-specific exon I.1 that mediates placental cell-specific expression. Using transgenic mice, we also observed that as little as 278 bp of DNA flanking the 5'-end of ovary-specific hCYP19 exon IIa was sufficient to target ovary-specific expression. This ovary-specific promoter contains response elements that bind cAMP-response element-binding protein (CREB) and the orphan nuclear receptors SF-1 and LRH-1, which are required for cAMP-mediated stimulation of CYP19 expression in granulosa and luteal cells during the estrous cycle and pregnancy. In this article, we review our studies to define genomic regions and response elements that mediate placenta-specific expression of the hCYP19 gene. The temporal and spatial expression of LRH-1 versus SF-1 in the developing gonad during mouse embryogenesis and in the postnatal ovary also will be considered.
Collapse
Affiliation(s)
- Carole R Mendelson
- Department of Biochemistry, The University of Texas Southwestern Medical Center at Dallas, TX 75390-9038, USA.
| | | |
Collapse
|
21
|
Wenzel PL, Wu L, de Bruin A, Chong JL, Chen WY, Dureska G, Sites E, Pan T, Sharma A, Huang K, Ridgway R, Mosaliganti K, Sharp R, Machiraju R, Saltz J, Yamamoto H, Cross JC, Robinson ML, Leone G. Rb is critical in a mammalian tissue stem cell population. Genes Dev 2007; 21:85-97. [PMID: 17210791 PMCID: PMC1759903 DOI: 10.1101/gad.1485307] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The inactivation of the retinoblastoma (Rb) tumor suppressor gene in mice results in ectopic proliferation, apoptosis, and impaired differentiation in extraembryonic, neural, and erythroid lineages, culminating in fetal death by embryonic day 15.5 (E15.5). Here we show that the specific loss of Rb in trophoblast stem (TS) cells, but not in trophoblast derivatives, leads to an overexpansion of trophoblasts, a disruption of placental architecture, and fetal death by E15.5. Despite profound placental abnormalities, fetal tissues appeared remarkably normal, suggesting that the full manifestation of fetal phenotypes requires the loss of Rb in both extraembryonic and fetal tissues. Loss of Rb resulted in an increase of E2f3 expression, and the combined ablation of Rb and E2f3 significantly suppressed Rb mutant phenotypes. This rescue appears to be cell autonomous since the inactivation of Rb and E2f3 in TS cells restored placental development and extended the life of embryos to E17.5. Taken together, these results demonstrate that loss of Rb in TS cells is the defining event causing lethality of Rb(-/-) embryos and reveal the convergence of extraembryonic and fetal functions of Rb in neural and erythroid development. We conclude that the Rb pathway plays a critical role in the maintenance of a mammalian stem cell population.
Collapse
Affiliation(s)
- Pamela L. Wenzel
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Lizhao Wu
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Alain de Bruin
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Jean-Leon Chong
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Wen-Yi Chen
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Geoffrey Dureska
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Emily Sites
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Tony Pan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Ashish Sharma
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Kun Huang
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Randall Ridgway
- Department of Computer Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Kishore Mosaliganti
- Department of Computer Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Richard Sharp
- Department of Computer Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Raghu Machiraju
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Computer Science and Engineering, The Ohio State University, Columbus, Ohio 43210, USA
| | - Joel Saltz
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Biomedical Informatics, Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| | - Hideyuki Yamamoto
- Department of Biochemistry and Molecular Biology, University of Calgary Faculty of Medicine, Calgary, Alberta T2N 4N1, Canada
| | - James C. Cross
- Department of Biochemistry and Molecular Biology, University of Calgary Faculty of Medicine, Calgary, Alberta T2N 4N1, Canada
| | - Michael L. Robinson
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Division of Molecular and Human Genetics, Children’s Research Institute, Columbus, Ohio 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio 43210, USA
- E-MAIL ; FAX (513) 529-6900
| | - Gustavo Leone
- Human Cancer Genetics Program, Department of Molecular Virology, Immunology and Medical Genetics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Molecular Genetics, College of Biological Sciences, The Ohio State University, Columbus, Ohio 43210, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
- Corresponding authors.E-MAIL ; FAX (614) 292-3312
| |
Collapse
|
22
|
Jiang B, Mendelson CR. O2 enhancement of human trophoblast differentiation and hCYP19 (aromatase) gene expression are mediated by proteasomal degradation of USF1 and USF2. Mol Cell Biol 2005; 25:8824-33. [PMID: 16199862 PMCID: PMC1265767 DOI: 10.1128/mcb.25.20.8824-8833.2005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2005] [Revised: 03/31/2005] [Accepted: 07/25/2005] [Indexed: 11/20/2022] Open
Abstract
When cultured in 20% O(2), human cytotrophoblasts fuse to form the syncytiotrophoblast with marked induction of hCYP19 (aromatase) gene expression. When cultured in 2% O(2), cytotrophoblast fusion and induced hCYP19 expression are prevented. These effects of hypoxia are mediated by increased expression of mammalian achaete/scute homologue-2 (Mash-2), which increases levels of upstream stimulatory factors 1 and 2 (USF1/2) and their binding as heterodimers to E-boxes surrounding the hCYP19 promoter. In studies to define mechanisms for O(2) regulation of syncytiotrophoblast differentiation, we found that hypoxia and overexpression of Mash-2 markedly increased cyclin B1 levels in cultured trophoblasts and the proportion of cells at the G(2)/M transition. Unlike USF proteins, USF1/2 mRNA levels are unaffected by O(2) tension. To determine whether increased O(2) might enhance proteasomal degradation of USF1/2, human trophoblasts were cultured in 2% or 20% O(2) with or without proteasome inhibitors. In cells cultured in 20% O(2), proteasome inhibitors increased USF1/2 protein levels and blocked spontaneous induction of hCYP19 expression, cell fusion, and differentiation. Like hypoxia, inhibitory effects of proteasome inhibitors on hCYP19 expression were mediated by increased binding of USF1/2 to the E-boxes. In human trophoblast cells cultured in 20% O(2), increased polyubiquitylation of USF1/2 proteins was observed. Thus, early in gestation when the placenta is relatively hypoxic, increased USF1/2 may block trophoblast differentiation and hCYP19 gene expression. In the second trimester, increased O(2) tension promotes proteasomal degradation of USF1/2, resulting in syncytiotrophoblast differentiation and induction of hCYP19 expression.
Collapse
Affiliation(s)
- Bing Jiang
- Departments of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, 75390, USA
| | | |
Collapse
|