1
|
Foyle KL, Chin PY, Merkwirth C, Wilson J, Hosking SL, Green ES, Chong MY, Zhang B, Moldenhauer LM, Ferguson GD, Morris GP, Karras JG, Care AS, Robertson SA. IL-2 Complexed With Anti-IL-2 Antibody Expands the Maternal T-Regulatory Cell Pool and Alleviates Fetal Loss in Abortion-Prone Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:2128-2149. [PMID: 39117109 DOI: 10.1016/j.ajpath.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Regulatory T (Treg) cells are essential for immune tolerance of embryo implantation, and insufficient Treg cells provokes early pregnancy loss. An abortion-prone mouse model was used to evaluate IL-2 complexed with JES6-1 anti-IL-2 antibody (IL-2/JES6-1) to boost uterine Treg cells and improve reproductive success. IL-2/JES6-1, but not IL-2/IgG, administered in periconception to CBA/J females mated with DBA/2 males elicited a greater than twofold increase in the proportion of CD4+ T cells expressing forkhead box P3 (FOXP3), and an increased ratio of FOXP3+ Treg cells/FOXP3- T conventional cells in the uterus and its draining lymph nodes at embryo implantation that was sustained into midgestation. An attenuated phenotype was evident in both thymic-derived and peripheral Treg cells with elevated cytotoxic T-lymphocyte antigen-4, CD25, and FOXP3 indicating improved suppressive function, as well as increased proliferative marker Ki-67. IL-2/JES6-1 treatment reduced fetal loss from 31% to 10%, accompanied by a 6% reduction in late gestation fetal weight, despite comparable placental size and architecture. Similar effects of IL-2/JES6-1 on Treg cells and fetal growth were seen in CBA/J females with healthy pregnancies sired by BALB/c males. These findings show that expanding the uterine Treg cell pool through targeting IL-2 signaling is a strategy worthy of further investigation for mitigating risk of immune-mediated fetal loss.
Collapse
Affiliation(s)
- Kerrie L Foyle
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Peck Y Chin
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | | | - Jasmine Wilson
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Shanna L Hosking
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Ella S Green
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Mei Y Chong
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Bihong Zhang
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Lachlan M Moldenhauer
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | | | - Gerald P Morris
- Department of Pathology, University of California, San Diego, La Jolla, California
| | | | - Alison S Care
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Sarah A Robertson
- The Robinson Research Institute and School of Biomedicine, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
2
|
Hercus JC, Salcedo Rubio DA, Osorio Nieto ME, Sturn MML, Keum C, Christians JK. The whole is lesser than the sum of its parts? Dissecting layer-enriched samples of rodent placenta is worth the effort. Placenta 2024:S0143-4004(24)00657-X. [PMID: 39317518 DOI: 10.1016/j.placenta.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/14/2024] [Accepted: 09/19/2024] [Indexed: 09/26/2024]
Abstract
Gene expression in the placenta, assessed by bulk RNA-seq, is a common method to explore placental function. Many rodent studies homogenize the entire placenta, and yet doing so may obscure differences within specific functional regions such as the labyrinth, junctional zone and decidua. Conversely, analysis of the whole placenta could generate apparent differences due to changes in composition (e.g., relative amounts of labyrinth vs junctional zone) rather than differential gene expression. We assess the value of dissecting and separately analysing the labyrinth and junctional zone/decidua by comparing RNA-seq results from the labyrinth, junctional zone/decidua combined, and whole placenta from an experiment examining effects of maternal food restriction and fetal sex in C57BL6/J mice at gestational day 17.5. The number of genes identified as differentially expressed in response to maternal food restriction was substantially higher in the labyrinth (910 genes), than in the junctional zone/decidua (50 genes), which in turn was slightly higher than in the whole placenta (3 genes). Only one gene was differentially expressed in all 3 tissue types, and 20 genes were differentially expressed in both the labyrinth and junctional zone/decidua. The larger number of differentially expressed genes in the labyrinth was due to both larger effect sizes and estimates of effect sizes having smaller standard errors. While dissection to obtain layer-enriched samples is slightly more time-consuming than collection of whole placenta and requires some practice, our results show that layer-enrichment is clearly worth the effort.
Collapse
Affiliation(s)
- Jess C Hercus
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada.
| | | | | | - Mackenzie M L Sturn
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada.
| | - Cheayeong Keum
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada.
| | - Julian K Christians
- Department of Biological Sciences, Simon Fraser University, Burnaby, British Columbia, Canada; Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada; British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada; Women's Health Research Institute, BC Women's Hospital and Health Centre, Vancouver, British Columbia, Canada.
| |
Collapse
|
3
|
Khoshkerdar A, Eid N, Batra V, Baker N, Holmes N, Henson S, Sang F, Wright V, McLaren J, Shakesheff K, Woad KJ, Morgan HL, Watkins AJ. Sub-Optimal Paternal Diet at the Time of Mating Disrupts Maternal Adaptations to Pregnancy in the Late Gestation Mouse. Nutrients 2024; 16:1879. [PMID: 38931234 PMCID: PMC11206308 DOI: 10.3390/nu16121879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Pregnancy represents a stage during which maternal physiology and homeostatic regulation undergo dramatic change and adaptation. The fundamental purpose of these adaptations is to ensure the survival of her offspring through adequate nutrient provision and an environment that is tolerant to the semi-allogenic foetus. While poor maternal diet during pregnancy is associated with perturbed maternal adaptations during pregnancy, the influence of paternal diet on maternal well-being is less clearly defined. We fed C57BL/6 male mice either a control (CD), low protein diet (LPD), a high fat/sugar Western diet (WD) or the LPD or WD supplemented with methyl donors (MD-LPD and MD-WD, respectively) for a minimum of 8 weeks prior to mating with C57BL/6 females. Mated females were culled at day 17 of gestation for the analysis of maternal metabolic, gut, cardiac and bone health. Paternal diet had minimal influences on maternal serum and hepatic metabolite levels or gut microbiota diversity. However, analysis of the maternal hepatic transcriptome revealed distinct profiles of differential gene expression in response to the diet of the father. Paternal LPD and MD-LPD resulted in differential expression of genes associated with lipid metabolism, transcription, ubiquitin conjugation and immunity in dams, while paternal WD and MD-WD modified the expression of genes associated with ubiquitin conjugation and cardiac morphology. Finally, we observed changes in maternal femur length, volume of trabecular bone, trabecular connectivity, volume of the cortical medullar cavity and thickness of the cortical bone in response to the father's diets. Our current study demonstrates that poor paternal diet at the time of mating can influence the patterns of maternal metabolism and gestation-associated adaptations to her physiology.
Collapse
Affiliation(s)
- Afsaneh Khoshkerdar
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Nader Eid
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Vipul Batra
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Nichola Baker
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Nadine Holmes
- Deep Seq, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (N.H.); (S.H.); (F.S.); (V.W.)
| | - Sonal Henson
- Deep Seq, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (N.H.); (S.H.); (F.S.); (V.W.)
| | - Fei Sang
- Deep Seq, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (N.H.); (S.H.); (F.S.); (V.W.)
| | - Victoria Wright
- Deep Seq, School of Life Sciences, Queen’s Medical Centre, University of Nottingham, Nottingham NG7 2UH, UK; (N.H.); (S.H.); (F.S.); (V.W.)
| | - Jane McLaren
- Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham NG7 2UH, UK; (J.M.)
| | - Kevin Shakesheff
- Regenerative Medicine and Cellular Therapies, School of Pharmacy, University of Nottingham, Nottingham NG7 2UH, UK; (J.M.)
| | - Kathryn J. Woad
- School of Veterinary Medicine and Science, University of Nottingham, Loughborough LE12 5RD, UK;
| | - Hannah L. Morgan
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| | - Adam J. Watkins
- Lifespan and Population Health, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK; (A.K.); (N.E.); (V.B.); (N.B.); (H.L.M.)
| |
Collapse
|
4
|
Candia AA, Lean SC, Zhang CXW, McKeating DR, Cochrane A, Gulacsi E, Herrera EA, Krause BJ, Sferruzzi-Perri AN. Obesogenic Diet in Mice Leads to Inflammation and Oxidative Stress in the Mother in Association with Sex-Specific Changes in Fetal Development, Inflammatory Markers and Placental Transcriptome. Antioxidants (Basel) 2024; 13:411. [PMID: 38671859 PMCID: PMC11047652 DOI: 10.3390/antiox13040411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Obesity during pregnancy is related to adverse maternal and neonatal outcomes. Factors involved in these outcomes may include increased maternal insulin resistance, inflammation, oxidative stress, and nutrient mishandling. The placenta is the primary determinant of fetal outcomes, and its function can be impacted by maternal obesity. The aim of this study on mice was to determine the effect of obesity on maternal lipid handling, inflammatory and redox state, and placental oxidative stress, inflammatory signaling, and gene expression relative to female and male fetal growth. METHODS Female mice were fed control or obesogenic high-fat/high-sugar diet (HFHS) from 9 weeks prior to, and during, pregnancy. On day 18.5 of pregnancy, maternal plasma, and liver, placenta, and fetal serum were collected to examine the immune and redox states. The placental labyrinth zone (Lz) was dissected for RNA-sequencing analysis of gene expression changes. RESULTS the HFHS diet induced, in the dams, hepatic steatosis, oxidative stress (reduced catalase, elevated protein oxidation) and the activation of pro-inflammatory pathways (p38-MAPK), along with imbalanced circulating cytokine concentrations (increased IL-6 and decreased IL-5 and IL-17A). HFHS fetuses were asymmetrically growth-restricted, showing sex-specific changes in circulating cytokines (GM-CSF, TNF-α, IL-6 and IFN-γ). The morphology of the placenta Lz was modified by an HFHS diet, in association with sex-specific alterations in the expression of genes and proteins implicated in oxidative stress, inflammation, and stress signaling. Placental gene expression changes were comparable to that seen in models of intrauterine inflammation and were related to a transcriptional network involving transcription factors, LYL1 and PLAG1. CONCLUSION This study shows that fetal growth restriction with maternal obesity is related to elevated oxidative stress, inflammatory pathways, and sex-specific placental changes. Our data are important, given the marked consequences and the rising rates of obesity worldwide.
Collapse
Affiliation(s)
- Alejandro A. Candia
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile;
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile;
- Department for the Woman and Newborn Health Promotion, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Samantha C. Lean
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Cindy X. W. Zhang
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Daniel R. McKeating
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Anna Cochrane
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Edina Gulacsi
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| | - Emilio A. Herrera
- Pathophysiology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, Universidad de Chile, Santiago 7500922, Chile;
| | - Bernardo J. Krause
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile;
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK; (A.A.C.); (C.X.W.Z.); (D.R.M.); (A.C.); (E.G.)
| |
Collapse
|
5
|
Wang Y. Erdr1 Drives Macrophage Programming via Dynamic Interplay with YAP1 and Mid1. Immunohorizons 2024; 8:198-213. [PMID: 38392560 PMCID: PMC10916360 DOI: 10.4049/immunohorizons.2400004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024] Open
Abstract
Erythroid differentiation regulator 1 (Erdr1) is a stress-induced, widely expressed, highly conserved secreted factor found in both humans and mice. Erdr1 is linked with the Hippo-YAP1 signaling. Initially identified as an inducer of hemoglobin synthesis, Erdr1 emerged as a multifunctional protein, especially in immune cells. Although Erdr1 has been implicated in regulating T cells and NK cell function, its role in macrophage remains unclear. This study explored the function and mechanism of Erdr1 in macrophage inflammatory response. The data demonstrated that Erdr1 could promote anti-inflammatory cytokine production, a function that also has been reported by previous research. However, I found Erdr1 also could play a proinflammatory role. The function of Erdr1 in macrophages depends on its dose and cell density. I observed that Erdr1 expression was inhibited in M1 macrophages but was upregulated in M2 macrophages compared with unpolarized macrophages. I hypothesized that Erdr1 balances the inflammatory response by binding with distinct adaptors dependent on varying concentrations. Mechanistically, I demonstrated YAP1 and Mid1 as the two adaptor proteins of Erdr1. The Erdr1-YAP1 interaction promotes anti-inflammatory cytokine production when Erdr1 levels are elevated, whereas the Erdr1-Mid1 interaction induces proinflammatory cytokine production when Erdr1 levels are decreased. This study highlights the effects of Erdr1 on regulating cytokine production from polarized macrophages potentially by regulating YAP1 in the nonclassical Hippo pathway.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Microbiology and Immunology, University of Iowa, IA City, IA
| |
Collapse
|
6
|
Meng XL, Yuan PB, Wang XJ, Hang J, Shi XM, Zhao YY, Wei Y. The Proteome Landscape of Human Placentas for Monochorionic Twins with Selective Intrauterine Growth Restriction. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1246-1259. [PMID: 37121272 PMCID: PMC11082409 DOI: 10.1016/j.gpb.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/20/2023] [Accepted: 03/09/2023] [Indexed: 05/02/2023]
Abstract
In perinatal medicine, intrauterine growth restriction (IUGR) is one of the greatest challenges. The etiology of IUGR is multifactorial, but most cases are thought to arise from placental insufficiency. However, identifying the placental cause of IUGR can be difficult due to numerous confounding factors. Selective IUGR (sIUGR) would be a good model to investigate how impaired placentation affects fetal development, as the growth discordance between monochorionic twins cannot be explained by confounding genetic or maternal factors. Herein, we constructed and analyzed the placental proteomic profiles of IUGR twins and normal cotwins. Specifically, we identified a total of 5481 proteins, of which 233 were differentially expressed (57 up-regulated and 176 down-regulated) in IUGR twins. Bioinformatics analysis indicates that these differentially expressed proteins (DEPs) are mainly associated with cardiovascular system development and function, organismal survival, and organismal development. Notably, 34 DEPs are significantly enriched in angiogenesis, and diminished placental angiogenesis in IUGR twins has been further elaborately confirmed. Moreover, we found decreased expression of metadherin (MTDH) in the placentas of IUGR twins and demonstrated that MTDH contributes to placental angiogenesis and fetal growth in vitro. Collectively, our findings reveal the comprehensive proteomic signatures of placentas for sIUGR twins, and the DEPs identified may provide in-depth insights into the pathogenesis of placental dysfunction and subsequent impaired fetal growth.
Collapse
Affiliation(s)
- Xin-Lu Meng
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Peng-Bo Yuan
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Xue-Ju Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Jing Hang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China; Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing 100191, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing 100191, China; National Clinical Research Center for Obstetrics and Gynecology, Beijing 100191, China
| | - Xiao-Ming Shi
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Yang-Yu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China.
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
7
|
Sferruzzi‐Perri AN, Lopez‐Tello J, Salazar‐Petres E. Placental adaptations supporting fetal growth during normal and adverse gestational environments. Exp Physiol 2023; 108:371-397. [PMID: 36484327 PMCID: PMC10103877 DOI: 10.1113/ep090442] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022]
Abstract
NEW FINDINGS What is the topic of this review? How the placenta, which transports nutrients and oxygen to the fetus, may alter its support of fetal growth developmentally and with adverse gestational conditions. What advances does it highlight? Placental formation and function alter with the needs of the fetus for substrates for growth during normal gestation and when there is enhanced competition for substrates in species with multiple gestations or adverse gestational environments, and this is mediated by imprinted genes, signalling pathways, mitochondria and fetal sexomes. ABSTRACT The placenta is vital for mammalian development and a key determinant of life-long health. It is the interface between the mother and fetus and is responsible for transporting the nutrients and oxygen a fetus needs to develop and grow. Alterations in placental formation and function, therefore, have consequences for fetal growth and birthweight, which in turn determine perinatal survival and risk of non-communicable diseases for the offspring in later postnatal life. However, the placenta is not a static organ. As this review summarizes, research from multiple species has demonstrated that placental formation and function alter developmentally to the needs of the fetus for substrates for growth during normal gestation, as well as when there is greater competition for substrates in polytocous species and monotocous species with multiple gestations. The placenta also adapts in response to the gestational environment, integrating information about the ability of the mother to provide nutrients and oxygen with the needs of the fetus in that prevailing environment. In particular, placental structure (e.g. vascularity, surface area, blood flow, diffusion distance) and transport capacity (e.g. nutrient transporter levels and activity) respond to suboptimal gestational environments, namely malnutrition, obesity, hypoxia and maternal ageing. Mechanisms mediating developmentally and environmentally induced homeostatic responses of the placenta that help support normal fetal growth include imprinted genes, signalling pathways, subcellular constituents and fetal sexomes. Identification of these placental strategies may inform the development of therapies for complicated human pregnancies and advance understanding of the pathways underlying poor fetal outcomes and their consequences for health and disease risk.
Collapse
Affiliation(s)
- Amanda Nancy Sferruzzi‐Perri
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Jorge Lopez‐Tello
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Esteban Salazar‐Petres
- Centre for Trophoblast Research, Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Facultad de CienciasDepartamento de Ciencias Básicas, Universidad Santo TomásValdiviaChile
| |
Collapse
|
8
|
Ihirwe RG, Martel J, Rahimi S, Trasler J. Protective and sex-specific effects of moderate dose folic acid supplementation on the placenta following assisted reproduction in mice. FASEB J 2023; 37:e22677. [PMID: 36515682 PMCID: PMC10108070 DOI: 10.1096/fj.202201428r] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/02/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022]
Abstract
Epigenetic defects induced by assisted reproductive technologies (ART) have been suggested as a potential mechanism contributing to suboptimal placentation. Here, we hypothesize that ART perturbs DNA methylation (DNAme) and gene expression during early placenta development, leading to abnormal placental phenotypes observed at term. Since folic acid (FA) plays a crucial role in epigenetic regulation, we propose that FA supplementation can rescue ART-induced placental defects. Female mice were placed on a control diet (CD), a moderate 4-fold (FAS4) or high dose 10-fold (FAS10) FA-supplemented diet prior to ART and compared to a natural mating group. ART resulted in 41 and 28 differentially expressed genes (DEGs) in E10.5 female and male placentas, respectively. Many DEGs were implicated in early placenta development and associated with DNAme changes; a number clustered at known imprinting control regions (ICR). In females, FAS4 partially corrected alterations in gene expression while FAS10 showed evidence of male-biased adverse effects. DNAme and gene expression for five genes involved in early placentation (Phlda2, EphB2, Igf2, Peg3, L3mbtl1) were followed up in placentas from normal as well as delayed and abnormal embryos. Phlda2 and Igf2 expression levels were lowest after ART in placentas of female delayed embryos. Moreover, ART concomitantly reduced DNAme at the Kcnq1ot1 ICR which regulates Phlda2 expression; FAS4 partially improved DNAme in a sex-specific manner. In conclusion, ART-associated placental DNAme and transcriptome alterations observed at mid-gestation are sex-specific; they may help explain adverse placental phenotypes detected at term and are partially corrected by maternal moderate dose FA supplementation.
Collapse
Affiliation(s)
- Rita Gloria Ihirwe
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Josée Martel
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Sophia Rahimi
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jacquetta Trasler
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.,Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada.,Department of Pediatrics, McGill University, Montreal, Quebec, Canada.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Salazar-Petres E, Pereira-Carvalho D, Lopez-Tello J, Sferruzzi-Perri AN. Placental structure, function, and mitochondrial phenotype relate to fetal size in each fetal sex in mice†. Biol Reprod 2022; 106:1292-1311. [PMID: 35293971 PMCID: PMC9327737 DOI: 10.1093/biolre/ioac056] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/28/2022] [Accepted: 03/08/2022] [Indexed: 11/25/2022] Open
Abstract
Fetal growth depends on placental function, which requires energy from mitochondria. Here we investigated whether mitochondrial function in the placenta relates to the growth of the lightest and heaviest fetuses of each sex within the litter of mice. Placentas from the lightest and heaviest fetuses were taken to evaluate placenta morphology (stereology), mitochondrial energetics (high-resolution respirometry), mitochondrial regulators, nutrient transporters, hormone handling, and signaling pathways (qPCR and Western blotting). We found that mitochondrial complex I and II oxygen consumption rate was greater for placentas supporting the lightest female fetuses, although placental complex I abundance of the lightest females and complexes III and V of the lightest males were decreased compared to their heaviest counterparts. Expression of mitochondrial biogenesis (Nrf1) and fission (Drp1 and Fis1) genes was lower in the placenta from the lightest females, whilst biogenesis-related gene Tfam was greater in the placenta of the lightest male fetuses. In addition, placental morphology and steroidogenic gene (Cyp17a1 and Cyp11a1) expression were aberrant for the lightest females, but glucose transporter (Slc2a1) expression was lower in only the lightest males versus their heaviest counterparts. Differences in intra-litter placental phenotype were related to changes in the expression of hormone-responsive (androgen receptor) and metabolic signaling (AMPK, AKT, and PPARγ) pathways. Thus, in normal mouse pregnancy, placental structure, function, and mitochondrial phenotype are differentially responsive to the growth of the female and male fetus. This study may inform the design of sex-specific therapies for placental insufficiency and fetal growth abnormalities with life-long benefits for the offspring.
Collapse
Affiliation(s)
- Esteban Salazar-Petres
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Daniela Pereira-Carvalho
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Jorge Lopez-Tello
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| | - Amanda Nancy Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Taraschi A, Cimini C, Colosimo A, Ramal-Sanchez M, Moussa F, Mokh S, Valbonetti L, Capacchietti G, Tagaram I, Bernabò N, Barboni B. Human Immune System Diseasome Networks and Female Oviductal Microenvironment: New Horizons to be Discovered. Front Genet 2022; 12:795123. [PMID: 35154249 PMCID: PMC8829125 DOI: 10.3389/fgene.2021.795123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022] Open
Abstract
Human hypofertility and infertility are two worldwide conditions experiencing nowadays an alarming increase due to a complex ensemble of events. The immune system has been suggested as one of the responsible for some of the etiopathogenic mechanisms involved in these conditions. To shed some light into the strong correlation between the reproductive and immune system, as can be inferred by the several and valuable manuscripts published to date, here we built a network using a useful bioinformatic tool (DisGeNET), in which the key genes involved in the sperm-oviduct interaction were linked. This constitutes an important event related with Human fertility since this interaction, and specially the spermatozoa, represents a not-self entity immunotolerated by the female. As a result, we discovered that some proteins involved in the sperm-oviduct interaction are implicated in several immune system diseases while, at the same time, some immune system diseases could interfere by using different pathways with the reproduction process. The data presented here could be of great importance to understand the involvement of the immune system in fertility reduction in Humans, setting the basis for potential immune therapeutic tools in the near future.
Collapse
Affiliation(s)
- Angela Taraschi
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise “G. Caporale”, Teramo, Italy
| | - Costanza Cimini
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Alessia Colosimo
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Marina Ramal-Sanchez
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Fadl Moussa
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Doctoral School of Science and Technology Lebanese University, Beirut, Lebanon
| | - Samia Mokh
- National Council for Scientific Research (CNRS), Lebanese Atomic Energy Commission (LAEC), Laboratory for Analysis of Organic Compound (LACO), Beiru, Lebanon
| | - Luca Valbonetti
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), National Research Council, Rome, Italy
| | - Giulia Capacchietti
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Israiel Tagaram
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Nicola Bernabò
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Institute of Biochemistry and Cell Biology (CNR-IBBC/EMMA/Infrafrontier/IMPC), National Research Council, Rome, Italy
- *Correspondence: Nicola Bernabò,
| | - Barbara Barboni
- Faculty of Biosciences and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
11
|
Aykroyd BRL, Tunster SJ, Sferruzzi-Perri AN. Loss of imprinting of the Igf2-H19 ICR1 enhances placental endocrine capacity via sex-specific alterations in signalling pathways in the mouse. Development 2022; 149:dev199811. [PMID: 34982814 PMCID: PMC8783045 DOI: 10.1242/dev.199811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 11/23/2021] [Indexed: 12/14/2022]
Abstract
Imprinting control region (ICR1) controls the expression of the Igf2 and H19 genes in a parent-of-origin specific manner. Appropriate expression of the Igf2-H19 locus is fundamental for normal fetal development, yet the importance of ICR1 in the placental production of hormones that promote maternal nutrient allocation to the fetus is unknown. To address this, we used a novel mouse model to selectively delete ICR1 in the endocrine junctional zone (Jz) of the mouse placenta (Jz-ΔICR1). The Jz-ΔICR1 mice exhibit increased Igf2 and decreased H19 expression specifically in the Jz. This was accompanied by an expansion of Jz endocrine cell types due to enhanced rates of proliferation and increased expression of pregnancy-specific glycoprotein 23 in the placenta of both fetal sexes. However, changes in the endocrine phenotype of the placenta were related to sexually-dimorphic alterations to the abundance of Igf2 receptors and downstream signalling pathways (Pi3k-Akt and Mapk). There was no effect of Jz-ΔICR1 on the expression of targets of the H19-embedded miR-675 or on fetal weight. Our results demonstrate that ICR1 controls placental endocrine capacity via sex-dependent changes in signalling.
Collapse
Affiliation(s)
| | | | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| |
Collapse
|
12
|
Diao L, Hierweger AM, Wieczorek A, Arck PC, Thiele K. Disruption of Glucocorticoid Action on CD11c + Dendritic Cells Favors the Generation of CD4 + Regulatory T Cells and Improves Fetal Development in Mice. Front Immunol 2021; 12:729742. [PMID: 34764952 PMCID: PMC8576435 DOI: 10.3389/fimmu.2021.729742] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022] Open
Abstract
A wealth of innate and adaptive immune cells and hormones are involved in mounting tolerance towards the fetus, a key aspect of successful reproduction. We could recently show that the specific cross talk between the pregnancy hormone progesterone and dendritic cells (DCs) is significantly engaged in the generation of CD4+ FoxP3+ regulatory T (Treg) cells while a disruption led to placental alterations and intra-uterine growth restriction. Apart from progesterone, also glucocorticoids affect immune cell functions. However, their functional relevance in the context of pregnancy still needs clarification. We developed a mouse line with a selective knockout of the glucocorticoid receptor (GR) on DCs, utilizing the cre/flox system. Reproductive outcome and maternal immune and endocrine adaptation of Balb/c-mated C57Bl/6 GRflox/floxCD11ccre/wt (mutant) females was assessed on gestation days (gd) 13.5 and 18.5. Balb/c-mated C57Bl/6 GRwt/wtCD11ccre/wt (wt) females served as controls. The number of implantation and fetal loss rate did not differ between groups. However, we identified a significant increase in fetal weight in fetuses from mutant dams. While the frequencies of CD11c+ cells remained largely similar, a decreased expression of co-stimulatory molecules was observed on DCs of mutant females on gd 13.5, along with higher frequencies of CD4+ and CD8+ Treg cells. Histomorphological and gene expression analysis revealed an increased placental volume and an improved functional placental capacity in mice lacking the GR on CD11c+ DCs. In summary, we here demonstrate that the disrupted communication between GCs and DCs favors a tolerant immune microenvironment and improves placental function and fetal development.
Collapse
Affiliation(s)
| | | | | | | | - Kristin Thiele
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
13
|
Van Gronigen Case G, Storey KM, Parmeley LE, Schulz LC. Effects of maternal nutrient restriction during the periconceptional period on placental development in the mouse. PLoS One 2021; 16:e0244971. [PMID: 33444393 PMCID: PMC7808591 DOI: 10.1371/journal.pone.0244971] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/20/2020] [Indexed: 01/13/2023] Open
Abstract
Maternal undernutrition has detrimental effects on fetal development and adult health. Total caloric restriction during early pregnancy followed by adequate nutrition for the remainder of gestation, is particularly linked to cardiovascular and metabolic disease risks during adulthood. The placenta is responsible for transport of nutrients from the maternal to fetal circulation, and the efficiency with which it does so can be adjusted to the maternal nutrient supply. There is evidence that placental adaptations to nutrient restriction in early pregnancy may be retained even when adequate nutrition is restored later in pregnancy, leading to a potential mismatch between placental efficiency and maternal nutrient supplies. However, in the mouse, 50% caloric restriction from days 1.5-11.5 of gestation, while temporarily altering placental structure and gene expression, had no significant effect on day 18.5. The periconceptional period, during which oocyte maturation, fertilization, and preimplantation development occur may be especially critical in creating lasting impact on the placenta. Here, mice were subjected to 50% caloric restriction from 3 weeks prior to pregnancy through d11.5, and then placental structure, the expression of key nutrient transporters, and global DNA methylation levels were examined at gestation d18.5. Prior exposure to caloric restriction increased maternal blood space area, but decreased expression of the key System A amino acid transporter Slc38a4 at d18.5. Neither placental and fetal weights, nor placental DNA methylation levels were affected. Thus, total caloric restriction beginning in the periconceptional period does have a lasting impact on placental development in the mouse, but without changing placental efficiency.
Collapse
Affiliation(s)
- Gerialisa Van Gronigen Case
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO, United States of America
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States of America
| | - Kathryn M. Storey
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO, United States of America
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States of America
| | - Lauren E. Parmeley
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO, United States of America
- Division of Biological Sciences, University of Missouri, Columbia, MO, United States of America
| | - Laura C. Schulz
- Department of Obstetrics, Gynecology and Women’s Health, University of Missouri, Columbia, MO, United States of America
| |
Collapse
|
14
|
Siqueira LG, Silva MVG, Panetto JC, Viana JH. Consequences of assisted reproductive technologies for offspring function in cattle. Reprod Fertil Dev 2020; 32:82-97. [PMID: 32188560 DOI: 10.1071/rd19278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abnormal fetuses, neonates and adult offspring derived by assisted reproductive technologies (ART) have been reported in humans, rodents and domestic animals. The use of ART has also been associated with an increased likelihood of certain adult diseases. These abnormalities may arise as a result of an excess of or missing maternally derived molecules during invitro culture, because the invitro environment is artificial and suboptimal for embryo development. Nonetheless, the success of ART in overcoming infertility or improving livestock genetics is undeniable. Limitations of invitro embryo production (IVEP) in cattle include lower rates of the establishment and maintenance of pregnancy and an increased incidence of neonatal morbidity and mortality. Moreover, recent studies demonstrated long-term effects of IVEP in cattle, including increased postnatal mortality, altered growth and a slight reduction in the performance of adult dairy cows. This review addresses the effects of an altered preimplantation environment on embryo and fetal programming and offspring development. We discuss cellular and molecular responses of the embryo to the maternal environment, how ART may disturb programming, the possible role of epigenetic effects as a mechanism for altered phenotypes and long-term effects of ART that manifest in postnatal life.
Collapse
Affiliation(s)
- Luiz G Siqueira
- Embrapa Gado de Leite, Juiz de Fora, MG, Brazil 36038-330; and Corresponding author.
| | | | - João C Panetto
- Embrapa Gado de Leite, Juiz de Fora, MG, Brazil 36038-330
| | - João H Viana
- Embrapa Recursos Genéticos e Biotecnologia, Brasília, DF, Brazil 70770-917
| |
Collapse
|
15
|
The BlastGen study: a randomized controlled trial of blastocyst media supplemented with granulocyte-macrophage colony-stimulating factor. Reprod Biomed Online 2020; 40:645-652. [PMID: 32220517 DOI: 10.1016/j.rbmo.2020.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/19/2019] [Accepted: 01/14/2020] [Indexed: 11/21/2022]
Abstract
RESEARCH QUESTION Does Embryogen®/BlastGen™ culture medium improve live birth rates compared with standard culture medium for women undergoing IVF and intracytoplasmic sperm injection (ICSI) with poor prognosis. DESIGN Randomized clinical trial. A total of 100 couples undergoing IVF/ICSI were randomly allocated to having their inseminated oocytes incubated in either Embryogen®/BlastGen™ sequential culture media or standard Cleavage/Blastocyst sequential culture media for 5 days (ClinicalTrials.gov Identifier: NCT02305420). RESULTS No statistically significant difference in live birth rate was found between the control group and the Embryogen®/BlastGen™ group (17 [34%] versus 11 [22%], respectively) (OR 0.55; 95% CI 0.22 to 1.32; P = 0.18). After adjustment for maternal age, body mass index and fertilization procedure, the blastulation rate reduced (40.6 ± 26.5 versus 24.6 ± 26.7; RR 0.70, CI 0.52 to 0.95; P < 0.05), and grade of the embryo transferred (OR 0.35, CI 0.16 to 0.77; P < 0.01) when Embryogen®/BlastGen™ medium was used. CONCLUSION A significant reduction in day-5 embryo outcome parameters was found using Embryogen®/BlastGen™ compared with standard medium, and insufficient evidence of a difference in pregnancy outcomes. Taking into consideration the small samples size, study limitations and strict inclusion criteria of this single-centre study, further research is needed to determine the efficacy of Embryogen®/BlastGen™ medium in couples undergoing IVF/ICSI.
Collapse
|
16
|
Napso T, Hung YP, Davidge ST, Care AS, Sferruzzi-Perri AN. Advanced maternal age compromises fetal growth and induces sex-specific changes in placental phenotype in rats. Sci Rep 2019; 9:16916. [PMID: 31780670 PMCID: PMC6882885 DOI: 10.1038/s41598-019-53199-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 10/29/2019] [Indexed: 12/21/2022] Open
Abstract
Advanced maternal age is associated with an increased risk of pregnancy complications. It programmes sex-specific cardiovascular dysfunction in rat offspring, however the intrauterine mechanisms involved remain unknown. This study in the rat assessed the impact of advanced maternal age on placental phenotype in relation to the growth of female and male fetuses. We show that relative to young (3-4 months) dams, advanced maternal age (9.5-10 months) compromises growth of both female and male fetuses but affects the placental phenotype sex-specifically. In placentas from aged versus young dams, the size of the placental transport and endocrine zones were increased and expression of Igf2 (+41%) and placental lactogen (Prl3b1: +59%) genes were upregulated in female, but not male fetuses. Placental abundance of IGF2 protein also decreased in the placenta of males only (-95%). Moreover, in placentas from aged versus young dams, glucocorticoid metabolism (11β-hsd2: +63% and 11β-hsd1: -33%) was higher in females, but lower in males (11β-hsd2: -50% and 11β-hsd1: unaltered). There was however, no change in the placental abundance of 11β-HSD2 protein in aged versus young dams regardless of fetal sex. Levels of oxidative stress in the placenta were increased in female and male fetuses (+57% and +90%, respectively) and apoptosis increased specifically in the placenta of males from aged rat dams (+700%). Thus, advanced maternal age alters placental phenotype in a sex-specific fashion. These sexually-divergent changes may play a role in determining health outcomes of female and male offspring of aged mothers.
Collapse
Affiliation(s)
- Tina Napso
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Yin-Po Hung
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sandra T Davidge
- Department of Obstetrics and Gynaecology, Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Alison S Care
- Department of Obstetrics and Gynaecology, Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
- Robinson Research Institute and Adelaide Medical School, University of Adelaide, South Australia, Australia
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
17
|
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: 1.0] [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
|
18
|
Thiele K, Hierweger AM, Riquelme JIA, Solano ME, Lydon JP, Arck PC. Impaired Progesterone-Responsiveness of CD11c + Dendritic Cells Affects the Generation of CD4 + Regulatory T Cells and Is Associated With Intrauterine Growth Restriction in Mice. Front Endocrinol (Lausanne) 2019; 10:96. [PMID: 30858825 PMCID: PMC6397849 DOI: 10.3389/fendo.2019.00096] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/01/2019] [Indexed: 12/13/2022] Open
Abstract
Up to 10% of pregnancies in Western societies are affected by intrauterine growth restriction (IUGR). IUGR reduces short-term neonatal survival and impairs long-term health of the children. To date, the molecular mechanisms involved in the pathogenesis of IUGR are largely unknown, but the failure to mount an adequate endocrine and immune response during pregnancy has been proposed to facilitate the occurrence of IUGR. A cross talk between the pregnancy hormone progesterone and innate immune cell subsets such as dendritic cells (DCs) is vital to ensure adequate placentation and fetal growth. However, experimental strategies to pinpoint distinct immune cell subsets interacting with progesterone in vivo have long been limited. In the present study, we have overcome this limitation by generating a mouse line with a specific deletion of the progesterone receptor (PR) on CD11c+ DCs. We took advantage of the cre/loxP system and assessed reproductive outcome in Balb/c-mated C57Bl/6 PRflox/floxCD11ccre/wt females. Balb/c-mated C57Bl/6 PRwt/wtCD11ccre/wt females served as controls. In all dams, fetal growth and development, placental function and maternal immune and endocrine adaptation were evaluated at different gestational time points. We observed a significantly reduced fetal weight on gestational day 13.5 and 18.5 in PRflox/floxCD11ccre/wt females. While frequencies of uterine CD11c+ cells were similar in both groups, an increased frequency of co-stimulatory molecules was observed on DCs in PRflox/floxCD11ccre/wt mice, along with reduced frequencies of CD4+ FoxP3+ and CD8+ CD122+ regulatory T (Treg) cells. Placental histomorphology revealed a skew toward increased junctional zone at the expense of the labyrinth in implantations of PRflox/floxCD11ccre/wt females, accompanied by increased plasma progesterone concentrations. Our results support that DCs are highly responsive to progesterone, subsequently adapting to a tolerogenic phenotype. If such cross talk between progesterone and DCs is impaired, the generation of pregnancy-protective immune cells subsets such as CD4+ and CD8+ Treg cells is reduced, which is associated with poor placentation and IUGR in mice.
Collapse
Affiliation(s)
- Kristin Thiele
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- *Correspondence: Kristin Thiele
| | - Alexandra Maximiliane Hierweger
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Isabel Amambay Riquelme
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - María Emilia Solano
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - John P. Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Petra Clara Arck
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Petra Clara Arck
| |
Collapse
|
19
|
Shi GD, Zhang XL, Cheng X, Wang X, Fan BY, Liu S, Hao Y, Wei ZJ, Zhou XH, Feng SQ. Abnormal DNA Methylation in Thoracic Spinal Cord Tissue Following Transection Injury. Med Sci Monit 2018; 24:8878-8890. [PMID: 30531681 PMCID: PMC6295140 DOI: 10.12659/msm.913141] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Spinal cord injury (SCI) is a serious disease with high disability and mortality rates, with no effective therapeutic strategies available. In SCI, abnormal DNA methylation is considered to be associated with axonal regeneration and cell proliferation. However, the roles of key genes in potential molecular mechanisms of SCI are not clear. Material/Methods Subacute spinal cord injury models were established in Wistar rats. Histological observations and motor function assessments were performed separately. Whole-genome bisulfite sequencing (WGBS) was used to detect the methylation of genes. Gene ontology (GO) term enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed using the DAVID database. Protein–protein interaction (PPI) networks were analyzed by Cytoscape software. Results After SCI, many cavities, areas of necrotic tissue, and many inflammatory cells were observed, and motor function scores were low. After the whole-genome bisulfite sequencing, approximately 96 DMGs were screened, of which 50 were hypermethylated genes and 46 were hypomethylated genes. KEGG pathway analysis highlighted the Axon Guidance pathway, Endocytosis pathway, T cell receptor signaling pathway, and Hippo signaling pathway. Expression patterns of hypermethylated genes and hypomethylated genes detected by qRT-PCR were the opposite of WGBS data, and the difference was significant. Conclusions Abnormal methylated genes and key signaling pathways involved in spinal cord injury were identified through histological observation, behavioral assessment, and bioinformatics analysis. This research can serve as a source of additional information to expand understanding of spinal cord-induced epigenetic changes.
Collapse
Affiliation(s)
- Gui-Dong Shi
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Xiao-Lei Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Xin Cheng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Xu Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Bao-You Fan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Shen Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Yan Hao
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Zhi-Jian Wei
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Xian-Hu Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| | - Shi-Qing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China (mainland).,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China (mainland)
| |
Collapse
|
20
|
Furukawa S, Tsuji N, Sugiyama A. Morphology and physiology of rat placenta for toxicological evaluation. J Toxicol Pathol 2018; 32:1-17. [PMID: 30739991 PMCID: PMC6361663 DOI: 10.1293/tox.2018-0042] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/04/2018] [Indexed: 12/18/2022] Open
Abstract
The placenta plays a pivotal role in fetal growth, and placental dysfunction and injury are associated with embryo/fetal toxicity. Histological examination of the rat placenta for safety evaluation provides valuable clues to the mechanisms of this toxicity. However, the placenta has specific and complex biological features unlike those of other organs, and placental structure dramatically changes depending on the time during the gestation period. Thus, time-dependent histopathological examination of the rat placenta should be performed based on the understanding of normal developmental changes in morphology and function. The placentas of rats and humans are both anatomically classified as discoid and hemochorial types. However, there are differences between rats and humans in terms of placental histological structure, the fetal-maternal interface, and the function of the yolk sac. Therefore, extrapolation of placental toxicity from rats to humans should be done cautiously in the evaluation of risk factors. This review describes the development, morphology, physiology, and toxicological features of the rat placenta and the differences between the rat and human placenta to enable accurate evaluation of reproductive and developmental toxicity in studies.
Collapse
Affiliation(s)
- Satoshi Furukawa
- Biological Research Laboratories, Nissan Chemical Corporation, 1470 Shiraoka, Shiraoka-shi, Saitama 349-0294, Japan
| | - Naho Tsuji
- Biological Research Laboratories, Nissan Chemical Corporation, 1470 Shiraoka, Shiraoka-shi, Saitama 349-0294, Japan
| | - Akihiko Sugiyama
- Veterinary Clinical Pathology, Faculty of Veterinary Medicine Okayama University of Science, 1-3 Ikoinooka, Imabari, Ehime794-8555, Japan
| |
Collapse
|
21
|
Remus CC, Kording F, Arck P, Solano E, Sedlacik J, Adam G, Hecher K, Forkert ND. DCE MRI reveals early decreased and later increased placenta perfusion after a stress challenge during pregnancy in a mouse model. Placenta 2018; 65:15-19. [PMID: 29908637 DOI: 10.1016/j.placenta.2018.03.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 01/27/2023]
Abstract
OBJECTIVES Stress during pregnancy is known to have negative effects on fetal outcome. The purpose of this exploratory study was to examine placental perfusion alterations after stress challenge during pregnancy in a mouse model. MATERIAL AND METHODS Seven Tesla MRI was performed on pregnant mice at embrionic day (ED) 14.5 and 16.5. Twenty dams were exposed to an established acoustic stress challenge model while twenty non-exposed dams served as controls. Placental perfusion was analyzed in dynamic contrast-enhanced (DCE) MRI using the steepest slope model. The two functional placental compartments, the highly vascularized labyrinth and the endocrine junctional zone, were assessed separately. RESULTS Statistical analysis revealed decreased perfusion levels in the stress group at ED 14.5 compared to controls in both placenta compartments. On ED 16.5, the perfusion level increased significantly in the stress group while placenta perfusion in controls remained similar or even slightly decreased leading to an overall increased perfusion in the stress group on ED 16.5 compared to controls. CONCLUSION MR imaging allows noninvasive placenta perfusion assessment in this fetal stress mimicking animal model. In this exploratory study, we demonstrated that stress challenge during pregnancy leads to an initial reduction followed by an increase of placenta perfusion.
Collapse
Affiliation(s)
- Chressen Catharina Remus
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, Centre for Radiology and Endoscopy, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Fabian Kording
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, Centre for Radiology and Endoscopy, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Petra Arck
- Department of Obstetrics and Fetal Medicine, Center for Obstetrics and Paediatrics, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Emilia Solano
- Department of Obstetrics and Fetal Medicine, Center for Obstetrics and Paediatrics, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Jan Sedlacik
- Department of Diagnostic and Interventional Neuroradiology, Centre for Radiology and Endoscopy, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, Centre for Radiology and Endoscopy, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Kurt Hecher
- Department of Obstetrics and Fetal Medicine, Center for Obstetrics and Paediatrics, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Nils Daniel Forkert
- Department of Radiology and Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
22
|
Decato BE, Lopez-Tello J, Sferruzzi-Perri AN, Smith AD, Dean MD. DNA Methylation Divergence and Tissue Specialization in the Developing Mouse Placenta. Mol Biol Evol 2017; 34:1702-1712. [PMID: 28379409 DOI: 10.1093/molbev/msx112] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The placental epigenome plays a vital role in regulating mammalian growth and development. Aberrations in placental DNA methylation are linked to several disease states, including intrauterine growth restriction and preeclampsia. Studying the evolution and development of the placental epigenome is critical to understanding the origin and progression of such diseases. Although high-resolution studies have found substantial variation between placental methylomes of different species, the nature of methylome variation has yet to be characterized within any individual species. We conducted a study of placental DNA methylation at high resolution in multiple strains and closely related species of house mice (Mus musculus musculus, Mus m. domesticus, and M. spretus), across developmental timepoints (embryonic days 15-18), and between two distinct layers (labyrinthine transport and junctional endocrine). We observed substantial genome-wide methylation heterogeneity in mouse placenta compared with other differentiated tissues. Species-specific methylation profiles were concentrated in retrotransposon subfamilies, specifically RLTR10 and RLTR20 subfamilies. Regulatory regions such as gene promoters and CpG islands displayed cross-species conservation, but showed strong differences between layers and developmental timepoints. Partially methylated domains exist in the mouse placenta and widen during development. Taken together, our results characterize the mouse placental methylome as a highly heterogeneous and deregulated landscape globally, intermixed with actively regulated promoter and retrotransposon sequences.
Collapse
Affiliation(s)
- Benjamin E Decato
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA
| | - Jorge Lopez-Tello
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development, and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Andrew D Smith
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA
| | - Matthew D Dean
- Molecular and Computational Biology, Department of Biological Sciences, University of Southern California, Los Angeles, CA
| |
Collapse
|
23
|
Smith A, Witte E, McGee D, Knott J, Narang K, Racicot K. Cortisol inhibits CSF2 and CSF3 via DNA methylation and inhibits invasion in first-trimester trophoblast cells. Am J Reprod Immunol 2017; 78. [PMID: 28846166 DOI: 10.1111/aji.12741] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/20/2017] [Indexed: 12/22/2022] Open
Abstract
PROBLEM Heightened maternal stress affects trophoblast function and increases risk for adverse pregnancy outcomes. METHODS OF STUDY Studies were performed using the first-trimester trophoblast cell line, Sw.71. Cytokines were quantified using qPCR and ELISA. Epigenetic regulation of cytokines was characterized by inhibiting histone deacetylation (1 μmol/L suberoylanilide hydroxamic acid [SAHA]) or methylation (5 μmol/L 5-azacytidine), or with chromatin immunoprecipitation (ChIP) with a pan-acetyl histone-3 antibody. Invasion assays used Matrigel chambers. RESULTS Cortisol inhibited expression of CSF2 (GM-CSF) and CSF3 (G-CSF) in trophoblast cells. Cortisol-associated inhibition was dependent on DNA methylation and was not affected by acetylation. There was also a modest decrease in trophoblast invasion, not dependent on loss of CSFs. CONCLUSION In first-trimester trophoblast cells, the physiological glucocorticoid, cortisol, inhibited two cytokines with roles in placental development and decreased trophoblast invasion. Cortisol-associated changes in trophoblast function could increase the risk for immune-mediated abortion or other adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Arianna Smith
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Elizabeth Witte
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Devin McGee
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Jason Knott
- Department of Animal Science, College of Natural Resources, Michigan State University, East Lansing, MI, USA
| | - Kavita Narang
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.,Spectrum Health, Department of obstetrics and Gynecology, Grand Rapids, MI, USA
| | - Karen Racicot
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| |
Collapse
|
24
|
Potter EA, Dolgova EV, Proskurina AS, Efremov YR, Minkevich AM, Rozanov AS, Peltek SE, Nikolin VP, Popova NA, Seledtsov IA, Molodtsov VV, Zavyalov EL, Taranov OS, Baiborodin SI, Ostanin AA, Chernykh ER, Kolchanov NA, Bogachev SS. Gene expression profiling of tumor-initiating stem cells from mouse Krebs-2 carcinoma using a novel marker of poorly differentiated cells. Oncotarget 2017; 8:9425-9441. [PMID: 28031533 PMCID: PMC5354742 DOI: 10.18632/oncotarget.14116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/15/2016] [Indexed: 12/18/2022] Open
Abstract
Using the ability of poorly differentiated cells to natively internalize fragments of extracellular double-stranded DNA as a marker, we isolated a tumorigenic subpopulation present in Krebs-2 ascites that demonstrated the features of tumor-inducing cancer stem cells. Having combined TAMRA-labeled DNA probe and the power of RNA-seq technology, we identified a set of 168 genes specifically expressed in TAMRA-positive cells (tumor-initiating stem cells), these genes remaining silent in TAMRA-negative cancer cells. TAMRA+ cells displayed gene expression signatures characteristic of both stem cells and cancer cells. The observed expression differences between TAMRA+ and TAMRA- cells were validated by Real Time PCR. The results obtained corroborated the biological data that TAMRA+ murine Krebs-2 tumor cells are tumor-initiating stem cells. The approach developed can be applied to profile any poorly differentiated cell types that are capable of immanent internalization of double-stranded DNA.
Collapse
Affiliation(s)
- Ekaterina A. Potter
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Evgenia V. Dolgova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Anastasia S. Proskurina
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Yaroslav R. Efremov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
- Novosibirsk State University, Novosibirsk 630090, Russia
| | - Alexandra M. Minkevich
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Aleksey S. Rozanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sergey E. Peltek
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Valeriy P. Nikolin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Nelly A. Popova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
- Novosibirsk State University, Novosibirsk 630090, Russia
| | | | - Vladimir V. Molodtsov
- Novosibirsk State University, Novosibirsk 630090, Russia
- Softberry Inc., New York 10549, USA
| | - Evgeniy L Zavyalov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Oleg S. Taranov
- The State Research Center of Virology and Biotechnology VECTOR, Koltsovo, Novosibirsk 630559, Russia
| | - Sergey I. Baiborodin
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Alexander A. Ostanin
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Elena R. Chernykh
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Medical Sciences, Novosibirsk 630099, Russia
| | - Nikolay A. Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - Sergey S. Bogachev
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| |
Collapse
|
25
|
Sankar A, Kooistra SM, Gonzalez JM, Ohlsson C, Poutanen M, Helin K. Maternal expression of the JMJD2A/KDM4A histone demethylase is critical for pre-implantation development. Development 2017; 144:3264-3277. [DOI: 10.1242/dev.155473] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 08/14/2017] [Indexed: 12/20/2022]
Abstract
Regulation of chromatin composition through post-translational modifications of histones contributes to transcriptional regulation and is essential for many cellular processes, including differentiation and development. JMJD2A/KDM4A is a lysine demethylase with specificity towards di- and tri-methylated lysine 9 and lysine 36 of histone H3 (H3K9me2/me3 and H3K36me2/me3). Here, we report that Kdm4a as a maternal factor plays a key role in embryo survival and is vital for female fertility. Kdm4a−/- female mice ovulate normally with comparable fertilization but poor implantation rates, and cannot support healthy transplanted embryos to term. This is due to a role for Kdm4a in uterine function, where its loss causes reduced expression of key genes involved in ion transport, nutrient supply and cytokine signalling, that impact embryo survival. In addition, a significant proportion of Kdm4a deficient oocytes displays a poor intrinsic ability to develop into blastocysts. These embryos cannot compete with healthy embryos for implantation in vivo, highlighting Kdm4a as a maternal effect gene. Thus, our study dissects an important dual role for maternal Kdm4a in determining faithful early embryonic development and the implantation process.
Collapse
Affiliation(s)
- Aditya Sankar
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Denmark
- The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Present Address: Centre for Chromosome Stability, Institute of Cellular and Molecular Medicine, University of Copenhagen, Denmark
| | - Susanne Marije Kooistra
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Denmark
- Present Address: Department of Neuroscience, University Medical Centre, Groningen, University of Groningen, Groningen, The Netherlands
| | - Javier Martin Gonzalez
- Core Facility for Transgenic Mice, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Claes Ohlsson
- Department of Physiology Turku Center for Disease Modeling (TCDM), Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Poutanen
- Centre for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Physiology Turku Center for Disease Modeling (TCDM), Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kristian Helin
- Biotech Research and Innovation Centre, University of Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Denmark
- The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
26
|
Furukawa S, Tsuji N, Kobayashi Y, Yamagishi Y, Hayashi S, Abe M, Kuroda Y, Kimura M, Hayakawa C, Sugiyama A. Effect of dibutyltin on placental and fetal toxicity in rat. J Toxicol Sci 2017; 42:741-753. [DOI: 10.2131/jts.42.741] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Satoshi Furukawa
- Biological Research Laboratories, Nissan Chemical Industries, Ltd
| | - Naho Tsuji
- Biological Research Laboratories, Nissan Chemical Industries, Ltd
| | | | | | - Seigo Hayashi
- Biological Research Laboratories, Nissan Chemical Industries, Ltd
| | - Masayoshi Abe
- Biological Research Laboratories, Nissan Chemical Industries, Ltd
| | - Yusuke Kuroda
- Biological Research Laboratories, Nissan Chemical Industries, Ltd
| | - Masayuki Kimura
- Biological Research Laboratories, Nissan Chemical Industries, Ltd
| | - Chisato Hayakawa
- Biological Research Laboratories, Nissan Chemical Industries, Ltd
| | - Akihiko Sugiyama
- Courses of Veterinary Laboratory Medicine, School of Veterinary Medicine, Faculty of Agriculture, Tottori University
| |
Collapse
|
27
|
Siqueira LGB, Hansen PJ. Sex differences in response of the bovine embryo to colony-stimulating factor 2. Reproduction 2016; 152:645-654. [PMID: 27601717 PMCID: PMC5097130 DOI: 10.1530/rep-16-0336] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/05/2016] [Indexed: 01/09/2023]
Abstract
We tested whether gene expression of the bovine morula is modified by CSF2 in a sex-dependent manner and if sex determines the effect of CSF2 on competence of embryos to become blastocysts. Embryos were produced in vitro using X- or Y-sorted semen and treated at Day 5 of culture with 10 ng/mL bovine CSF2 or control. In experiment 1, morulae were collected at Day 6 and biological replicates (n = 8) were evaluated for transcript abundance of 90 genes by RT-qPCR using the Fluidigm Delta Gene assay. Expression of more than one-third (33 of 90) of genes examined was affected by sex. The effect of CSF2 on gene expression was modified by sex (P < 0.05) for five genes (DDX3Y/DDX3X-like, NANOG, MYF6, POU5F1 and RIPK3) and tended (P < 0.10) to be modified by sex for five other genes (DAPK1, HOXA5, PPP2R3A, PTEN and TNFSF8). In experiment 2, embryos were treated at Day 5 with control or CSF2 and blastocysts were collected at Day 7 for immunolabeling to determine the number of inner cell mass (ICM) and trophectoderm (TE) cells. CSF2 increased the percent of putative zygotes that became blastocysts for females, but did not affect the development of males. There was no effect of CSF2 or interaction of CSF2 with sex on the total number of blastomeres in blastocysts or in the number of inner cell mass or trophectoderm cells. In conclusion, CSF2 exerted divergent responses on gene expression and development of female and male embryos. These results are evidence of sexually dimorphic responses of the preimplantation embryo to this embryokine.
Collapse
Affiliation(s)
- Luiz G B Siqueira
- Department of Animal SciencesD.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, Florida, USA.,Embrapa Gado de LeiteJuiz de Fora, MG, Brazil
| | - Peter J Hansen
- Department of Animal SciencesD.H. Barron Reproductive and Perinatal Biology Research Program, and Genetics Institute, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
28
|
Godar RJ, Ma X, Liu H, Murphy JT, Weinheimer CJ, Kovacs A, Crosby SD, Saftig P, Diwan A. Repetitive stimulation of autophagy-lysosome machinery by intermittent fasting preconditions the myocardium to ischemia-reperfusion injury. Autophagy 2016; 11:1537-60. [PMID: 26103523 DOI: 10.1080/15548627.2015.1063768] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Autophagy, a lysosomal degradative pathway, is potently stimulated in the myocardium by fasting and is essential for maintaining cardiac function during prolonged starvation. We tested the hypothesis that intermittent fasting protects against myocardial ischemia-reperfusion injury via transcriptional stimulation of the autophagy-lysosome machinery. Adult C57BL/6 mice subjected to 24-h periods of fasting, every other day, for 6 wk were protected from in-vivo ischemia-reperfusion injury on a fed day, with marked reduction in infarct size in both sexes as compared with nonfasted controls. This protection was lost in mice heterozygous null for Lamp2 (coding for lysosomal-associated membrane protein 2), which demonstrate impaired autophagy in response to fasting with accumulation of autophagosomes and SQSTM1, an autophagy substrate, in the heart. In lamp2 null mice, intermittent fasting provoked progressive left ventricular dilation, systolic dysfunction and hypertrophy; worsening cardiomyocyte autophagosome accumulation and lack of protection to ischemia-reperfusion injury, suggesting that intact autophagy-lysosome machinery is essential for myocardial homeostasis during intermittent fasting and consequent ischemic cardioprotection. Fasting and refeeding cycles resulted in transcriptional induction followed by downregulation of autophagy-lysosome genes in the myocardium. This was coupled with fasting-induced nuclear translocation of TFEB (transcription factor EB), a master regulator of autophagy-lysosome machinery; followed by rapid decline in nuclear TFEB levels with refeeding. Endogenous TFEB was essential for attenuation of hypoxia-reoxygenation-induced cell death by repetitive starvation, in neonatal rat cardiomyocytes, in-vitro. Taken together, these data suggest that TFEB-mediated transcriptional priming of the autophagy-lysosome machinery mediates the beneficial effects of fasting-induced autophagy in myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Rebecca J Godar
- a Division of Cardiology and Center for Cardiovascular Research ; Department of Internal Medicine; Washington University School of Medicine ; St. Louis , MO USA.,b John Cochran VA Medical Center ; St. Louis , MO USA
| | - Xiucui Ma
- a Division of Cardiology and Center for Cardiovascular Research ; Department of Internal Medicine; Washington University School of Medicine ; St. Louis , MO USA.,b John Cochran VA Medical Center ; St. Louis , MO USA
| | - Haiyan Liu
- a Division of Cardiology and Center for Cardiovascular Research ; Department of Internal Medicine; Washington University School of Medicine ; St. Louis , MO USA
| | - John T Murphy
- a Division of Cardiology and Center for Cardiovascular Research ; Department of Internal Medicine; Washington University School of Medicine ; St. Louis , MO USA
| | - Carla J Weinheimer
- a Division of Cardiology and Center for Cardiovascular Research ; Department of Internal Medicine; Washington University School of Medicine ; St. Louis , MO USA
| | - Attila Kovacs
- a Division of Cardiology and Center for Cardiovascular Research ; Department of Internal Medicine; Washington University School of Medicine ; St. Louis , MO USA
| | - Seth D Crosby
- c Department of Genetics ; Washington University School of Medicine ; St. Louis , MO USA
| | - Paul Saftig
- d Institut für Biochemie; Christian-Albrechts-Universität zu Kiel ; Kiel , Germany
| | - Abhinav Diwan
- a Division of Cardiology and Center for Cardiovascular Research ; Department of Internal Medicine; Washington University School of Medicine ; St. Louis , MO USA.,b John Cochran VA Medical Center ; St. Louis , MO USA
| |
Collapse
|
29
|
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: 1.4] [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
|
30
|
Van Gronigen Caesar G, Dale JM, Osman EY, Garcia ML, Lorson CL, Schulz LC. Placental development in a mouse model of spinal muscular atrophy. Biochem Biophys Res Commun 2015; 470:82-87. [PMID: 26748185 DOI: 10.1016/j.bbrc.2015.12.120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/22/2015] [Indexed: 01/30/2023]
Abstract
Spinal Muscular Atrophy (SMA) is an autosomal recessive disorder, leading to fatal loss of motor neurons. It is caused by loss of function of the SMN gene, which is expressed throughout the body, and there is increasing evidence of dysfunction in non-neuronal tissues. Birthweight is one of most powerful prognostic factors for infants born with SMA, and intrauterine growth restriction is common. In the SMNΔ7 mouse model of SMA, pups with the disease lived 25% longer when their mothers were fed a higher fat, "breeder" diet. The placenta is responsible for transport of nutrients from mother to fetus, and is a major determinant of fetal growth. Thus, the present study tested the hypothesis that placental development is impaired in SMNΔ7 conceptuses. Detailed morphological characterization revealed no defects in SMNΔ7 placental development, and expression of key transcription factors regulating mouse placental development was unaffected. The intrauterine growth restriction observed in SMA infants likely does not result from impaired placental development.
Collapse
Affiliation(s)
- Gerialisa Van Gronigen Caesar
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA
| | - Jeffrey M Dale
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Erkan Y Osman
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
| | - Michael L Garcia
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| | - Christian L Lorson
- Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA; Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Laura C Schulz
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA; Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
31
|
Dutta S, Roy S, Polavaram NS, Stanton MJ, Zhang H, Bhola T, Hönscheid P, Donohue TM, Band H, Batra SK, Muders MH, Datta K. Neuropilin-2 Regulates Endosome Maturation and EGFR Trafficking to Support Cancer Cell Pathobiology. Cancer Res 2015; 76:418-28. [PMID: 26560516 DOI: 10.1158/0008-5472.can-15-1488] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 10/23/2015] [Indexed: 12/16/2022]
Abstract
Neuropilin-2 (NRP2) is a non-tyrosine kinase receptor frequently overexpressed in various malignancies, where it has been implicated in promoting many protumorigenic behaviors, such as imparting therapeutic resistance to metastatic cancer cells. Here, we report a novel function of NRP2 as a regulator of endocytosis, which is enhanced in cancer cells and is often associated with increased metastatic potential and drug resistance. We found that NRP2 depletion in human prostate and pancreatic cancer cells resulted in the accumulation of EEA1/Rab5-positive early endosomes concomitant with a decrease in Rab7-positive late endosomes, suggesting a delay in early-to-late endosome maturation. NRP2 depletion also impaired the endocytic transport of cell surface EGFR, arresting functionally active EGFR in endocytic vesicles that consequently led to aberrant ERK activation and cell death. Mechanistic investigations revealed that WD-repeat- and FYVE-domain-containing protein 1 (WDFY1) functioned downstream of NRP2 to promote endosome maturation, thereby influencing the endosomal trafficking of EGFR and the formation of autolysosomes responsible for the degradation of internalized cargo. Overall, our results indicate that the NRP2/WDFY1 axis is required for maintaining endocytic activity in cancer cells, which supports their oncogenic activities and confers drug resistance. Therefore, therapeutically targeting endocytosis may represent an attractive strategy to selectively target cancer cells in multiple malignancies.
Collapse
Affiliation(s)
- Samikshan Dutta
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sohini Roy
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Navatha S Polavaram
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Marissa J Stanton
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Heyu Zhang
- Department of Urologic Research, Biochemistry, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Tanvi Bhola
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Pia Hönscheid
- Institute of Pathology, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Terrence M Donohue
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska. Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska. Omaha VA Medical Center, Omaha, Nebraska
| | - Hamid Band
- Buffett Cancer Center, Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, Nebraska
| | - Surinder K Batra
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska. Buffett Cancer Center, Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, Nebraska
| | - Michael H Muders
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska. Institute of Pathology, University Hospital Carl Gustav Carus, TU, Dresden, Germany
| | - Kaustubh Datta
- Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska. Buffett Cancer Center, Eppley Cancer Institute, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
32
|
Solano ME, Kowal MK, O'Rourke GE, Horst AK, Modest K, Plösch T, Barikbin R, Remus CC, Berger RG, Jago C, Ho H, Sass G, Parker VJ, Lydon JP, DeMayo FJ, Hecher K, Karimi K, Arck PC. Progesterone and HMOX-1 promote fetal growth by CD8+ T cell modulation. J Clin Invest 2015; 125:1726-38. [PMID: 25774501 DOI: 10.1172/jci68140] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/29/2015] [Indexed: 12/20/2022] Open
Abstract
Intrauterine growth restriction (IUGR) affects up to 10% of pregnancies in Western societies. IUGR is a strong predictor of reduced short-term neonatal survival and impairs long-term health in children. Placental insufficiency is often associated with IUGR; however, the molecular mechanisms involved in the pathogenesis of placental insufficiency and IUGR are largely unknown. Here, we developed a mouse model of fetal-growth restriction and placental insufficiency that is induced by a midgestational stress challenge. Compared with control animals, pregnant dams subjected to gestational stress exhibited reduced progesterone levels and placental heme oxygenase 1 (Hmox1) expression and increased methylation at distinct regions of the placental Hmox1 promoter. These stress-triggered changes were accompanied by an altered CD8+ T cell response, as evidenced by a reduction of tolerogenic CD8+CD122+ T cells and an increase of cytotoxic CD8+ T cells. Using progesterone receptor- or Hmox1-deficient mice, we identified progesterone as an upstream modulator of placental Hmox1 expression. Supplementation of progesterone or depletion of CD8+ T cells revealed that progesterone suppresses CD8+ T cell cytotoxicity, whereas the generation of CD8+CD122+ T cells is supported by Hmox1 and ameliorates fetal-growth restriction in Hmox1 deficiency. These observations in mice could promote the identification of pregnancies at risk for IUGR and the generation of clinical interventional strategies.
Collapse
|
33
|
Brinkworth JF, Barreiro LB. The contribution of natural selection to present-day susceptibility to chronic inflammatory and autoimmune disease. Curr Opin Immunol 2014; 31:66-78. [PMID: 25458997 DOI: 10.1016/j.coi.2014.09.008] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/08/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022]
Abstract
Chronic inflammatory and autoimmune diseases have been the focus of many genome-wide association studies (GWAS) because they represent a significant cause of illness and morbidity, and many are heritable. Almost a decade of GWAS studies suggests that the pathological inflammation associated with these diseases is controlled by a limited number of networked immune system genes. Chronic inflammatory and autoimmune diseases are enigmatic from an evolutionary perspective because they exert a negative affect on reproductive fitness. The persistence of these conditions may be partially explained by the important roles the implicated immune genes play in pathogen defense and other functions thought to be under strong natural selection in humans. The evolutionary reasons for chronic inflammatory and autoimmune disease persistence and uneven distribution across populations are the focus of this review.
Collapse
Affiliation(s)
- Jessica F Brinkworth
- Sainte-Justine Hospital Research Centre, Montréal, Quebec H3T 1C5, Canada; Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Luis B Barreiro
- Sainte-Justine Hospital Research Centre, Montréal, Quebec H3T 1C5, Canada; Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada.
| |
Collapse
|
34
|
Identification of sites of STAT3 action in the female reproductive tract through conditional gene deletion. PLoS One 2014; 9:e101182. [PMID: 24983622 PMCID: PMC4077744 DOI: 10.1371/journal.pone.0101182] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/03/2014] [Indexed: 12/26/2022] Open
Abstract
The STAT3 transcription factor is a pleiotropic transducer of signalling by hormones, growth factors and cytokines that has been identified in the female reproductive tract from oocytes and granulosa cells of the ovary to uterine epithelial and stromal cells. In the present study we used transgenic models to investigate the importance of STAT3 for reproductive performance in these different tissues. The Cre-LoxP system was used to delete STAT3 in oocytes by crossing Stat3fl/fl with Zp3-cre+ mice, or in ovarian granulosa cells and uterine stroma by crossing with Amhr2-Cre+ mice. Surprisingly, deletion of STAT3 in oocytes had no effect on fertility indicating that the abundance of STAT3 protein in maturing oocytes and fertilized zygotes is not essential to these developmental stages. In Stat3fl/fl;Amhr2-cre+ females impaired fertility was observed through significantly fewer litters and smaller litter size. Ovulation rate, oocyte fertilization and development to blastocyst were unaffected in this line; however, poor recombination efficiency in granulosa cells had yielded no net change in STAT3 protein abundance. In contrast, uteri from these mice showed STAT3 protein depletion selectively from the stomal compartment. A significant reduction in number of viable fetuses on gestational day 18, increased fetal resorptions and disrupted placental morphology were evident causes of the reduced fertility. In conclusion, this study defines an important role for STAT3 in uterine stromal cells during embryo implantation and the development of a functional placenta.
Collapse
|
35
|
Rai A, Cross JC. Development of the hemochorial maternal vascular spaces in the placenta through endothelial and vasculogenic mimicry. Dev Biol 2014; 387:131-41. [PMID: 24485853 DOI: 10.1016/j.ydbio.2014.01.015] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/14/2014] [Accepted: 01/19/2014] [Indexed: 11/27/2022]
Abstract
The maternal vasculature within the placenta in primates and rodents is unique because it is lined by fetal cells of the trophoblast lineage and not by maternal endothelial cells. In addition to trophoblast cells that invade the uterine spiral arteries that bring blood into the placenta, other trophoblast subtypes sit at different levels of the vascular space. In mice, at least five distinct subtypes of trophoblast cells have been identified which engage maternal endothelial cells on the arterial and venous frontiers of the placenta, but which also form the channel-like spaces within it through a process analogous to formation of blood vessels (vasculogenic mimicry). These cells are all large, post-mitotic trophoblast giant cells. In addition to assuming endothelial cell-like characteristics (endothelial mimicry), they produce dozens of different hormones that are thought to regulate local and systemic maternal adaptations to pregnancy. Recent work has identified distinct molecular pathways in mice that regulate the morphogenesis of trophoblast cells on the arterial and venous sides of the vascular circuit that may be analogous to specification of arterial and venous endothelial cells.
Collapse
Affiliation(s)
- Anshita Rai
- Department of Biochemistry and Molecular Biology, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada; Department of Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1
| | - James C Cross
- Department of Biochemistry and Molecular Biology, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada; Department of Comparative Biology and Experimental Medicine, Obstetrics and Gynecology, and Medical Genetics, University of Calgary, HSC Room 2279, 3330 Hospital Drive NW, Calgary, Alta., Canada T2N 4N1.
| |
Collapse
|
36
|
Posey KL, Coustry F, Veerisetty AC, Liu P, Alcorn JL, Hecht JT. Chondrocyte-specific pathology during skeletal growth and therapeutics in a murine model of pseudoachondroplasia. J Bone Miner Res 2014; 29:1258-68. [PMID: 24194321 PMCID: PMC4075045 DOI: 10.1002/jbmr.2139] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 10/24/2013] [Accepted: 11/01/2013] [Indexed: 11/10/2022]
Abstract
Mutations in the gene encoding cartilage oligomeric matrix protein (COMP) cause pseudoachondroplasia (PSACH), a severe dwarfing condition. Pain, a significant complication, has generally been attributed to joint abnormalities and erosion and early onset osteoarthritis. Previously, we found that the inflammatory-related transcripts were elevated in growth plate and articular cartilages, indicating that inflammation plays an important role in the chondrocyte disease pathology and may contribute to the overall pain sequelae. Here, we describe the effects of D469-delCOMP expression on the skeleton and growth plate chondrocytes with the aim to define a treatment window and thereby reduce pain. Consistent with the human PSACH phenotype, skeletal development of D469del-COMP mice was normal and similar to controls at birth. By postnatal day 7 (P7), the D469del-COMP skeleton, limbs, skull and snout were reduced and this reduction was progressive during postnatal growth, resulting in a short-limbed dwarfed mouse. Modulation of prenatal and postnatal expression of D469del-COMP showed minimal retention/cell death at P7 with some retention/cell death by P14, suggesting that earlier treatment intervention at the time of PSACH diagnosis may produce optimal results. Important and novel findings were an increase in inflammatory proteins generally starting at P21 and that exercise exacerbates inflammation. These observations suggest that pain in PSACH may be related to an intrinsic inflammatory process that can be treated symptomatically and is not related to early joint erosion. We also show that genetic ablation of CHOP dampens the inflammatory response observed in mice expressing D469del-COMP. Toward identifying potential treatments, drugs known to decrease cellular stress (lithium, phenylbutyric acid, and valproate) were assessed. Interestingly, all diminished the chondrocyte pathology but had untoward outcomes on mouse growth, development, and longevity. Collectively, these results define an early treatment window in which chondrocytes can be salvaged, thereby potentially increasing skeletal growth and decreasing pain.
Collapse
Affiliation(s)
- Karen L Posey
- Department of Pediatrics, University of Texas Medical School at Houston, TX, USA
| | | | | | | | | | | |
Collapse
|
37
|
Mango RL, Wu QP, West M, McCook EC, Serody JS, van Deventer HW. C-C chemokine receptor 5 on pulmonary mesenchymal cells promotes experimental metastasis via the induction of erythroid differentiation regulator 1. Mol Cancer Res 2013; 12:274-82. [PMID: 24197118 DOI: 10.1158/1541-7786.mcr-13-0164] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
UNLABELLED C-C Chemokine receptor 5 knockout (Ccr5(-/-)) mice develop fewer experimental pulmonary metastases than wild-type (WT) mice. This phenomenon was explored by applying gene expression profiling to the lungs of mice with these metastases. Consequently, erythroid differentiation regulator 1 (Erdr1) was identified as upregulated in the WT mice. Though commonly associated with bone marrow stroma, Erdr1 was differentially expressed in WT pulmonary mesenchymal cells (PMC) and murine embryonic fibroblasts (MEF). Moreover, the Ccr5 ligand Ccl4 increased its expression by 3.36 ± 0.14-fold. Ccr5 signaling was dependent on the mitogen-activated protein kinase kinase (Map2k) but not the phosphoinositide 3-kinase (Pi3k) pathway because treatment with U0126 inhibited upregulation of Erdr1, but treatment with LY294002 increased the expression by 3.44 ± 0.92-fold (P < 0.05). The effect Erdr1 on B16-F10 melanoma metastasis was verified by the adoptive transfer of WT MEFs into Ccr5(-/-) mice. In this model, MEFs that had been transduced with Erdr1 short hairpin RNA (shRNA) lowered metastasis by 33% compared with control transduced MEFs. The relevance of ERDR1 on human disease was assessed by coculturing chronic lymphocytic leukemia (CLL) cells with M2-10B4 stromal cells that had been transfected with shRNA or control plasmids. After 96 hours of coculture, the cell counts were higher with control cell lines than with Erdr1 knockdown lines [odds ratio (OR), 1.88 ± 0.27, 2.52 ± 0.66, respectively]. This increase was associated with a decrease in apoptotic cells (OR, 0.69 ± 0.18, 0.58 ± 0.12, respectively). IMPLICATIONS Therefore, ERDR1 is a stromal-derived factor that promotes cancer cell survival in vitro and in an experimental metastasis model.
Collapse
Affiliation(s)
- Robert L Mango
- University of North Carolina, CB 7305, 170 Manning Drive, Chapel Hill, NC 27599-7305.
| | | | | | | | | | | |
Collapse
|
38
|
Sferruzzi-Perri AN, Vaughan OR, Haro M, Cooper WN, Musial B, Charalambous M, Pestana D, Ayyar S, Ferguson-Smith AC, Burton GJ, Constancia M, Fowden AL. An obesogenic diet during mouse pregnancy modifies maternal nutrient partitioning and the fetal growth trajectory. FASEB J 2013; 27:3928-37. [PMID: 23825226 DOI: 10.1096/fj.13-234823] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In developed societies, high-sugar and high-fat (HSHF) diets are now the norm and are increasing the rates of maternal obesity during pregnancy. In pregnant rodents, these diets lead to cardiovascular and metabolic dysfunction in their adult offspring, but the intrauterine mechanisms involved remain unknown. This study shows that, relative to standard chow, HSHF feeding throughout mouse pregnancy increases maternal adiposity (+30%, P<0.05) and reduces fetoplacental growth at d 16 (-10%, P<0.001). At d 19, however, HSHF diet group pup weight had normalized, despite the HSHF diet group placenta remaining small and morphologically compromised. This altered fetal growth trajectory was associated with enhanced placental glucose and amino acid transfer (+35%, P<0.001) and expression of their transporters (+40%, P<0.024). HSHF feeding also up-regulated placental expression of fatty acid transporter protein, metabolic signaling pathways (phosphoinositol 3-kinase and mitogen-activated protein kinase), and several growth regulatory imprinted genes (Igf2, Dlk1, Snrpn, Grb10, and H19) independently of changes in DNA methylation. Obesogenic diets during pregnancy, therefore, alter maternal nutrient partitioning, partly through changes in the placental phenotype, which helps to meet fetal nutrient demands for growth near term. However, by altering provision of specific nutrients, dietary-induced placental adaptations have important roles in programming development with health implications for the offspring in later life.
Collapse
Affiliation(s)
- Amanda N Sferruzzi-Perri
- 1Centre for Trophoblast Research, Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK CB2 3EG.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ziebe S, Loft A, Povlsen BB, Erb K, Agerholm I, Aasted M, Gabrielsen A, Hnida C, Zobel DP, Munding B, Bendz SH, Robertson SA. A randomized clinical trial to evaluate the effect of granulocyte-macrophage colony-stimulating factor (GM-CSF) in embryo culture medium for in vitro fertilization. Fertil Steril 2013; 99:1600-9. [DOI: 10.1016/j.fertnstert.2012.12.043] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 12/10/2012] [Accepted: 12/26/2012] [Indexed: 10/27/2022]
|
40
|
Rey Moreno MC, Fussell KC, Gröters S, Schneider S, Strauss V, Stinchcombe S, Fegert I, Veras M, van Ravenzwaay B. Epoxiconazole-Induced Degeneration in Rat Placenta and the Effects of Estradiol Supplementation. ACTA ACUST UNITED AC 2013; 98:208-21. [DOI: 10.1002/bdrb.21055] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 02/08/2013] [Indexed: 01/13/2023]
Affiliation(s)
| | - Karma C. Fussell
- Department of Experimental Toxicology and Ecology; BASF SE; Ludwigshafen; Germany
| | - Sibylle Gröters
- Department of Experimental Toxicology and Ecology; BASF SE; Ludwigshafen; Germany
| | - Steffen Schneider
- Department of Experimental Toxicology and Ecology; BASF SE; Ludwigshafen; Germany
| | - Volker Strauss
- Department of Experimental Toxicology and Ecology; BASF SE; Ludwigshafen; Germany
| | - Stefan Stinchcombe
- Department of Product Safety, Regulations, Toxicology and Ecology; BASF SE; Ludwigshafen; Germany
| | - Ivana Fegert
- Department of Product Safety, Regulations, Toxicology and Ecology; BASF SE; Ludwigshafen; Germany
| | - Mariana Veras
- LIM05-Department of Pathology; Laboratory of Environmental Air Pollution; University of Sao Paulo School of Medicine; Sao Paulo; Brazil
| | | |
Collapse
|
41
|
Schulz LC, Schlitt JM, Caesar G, Pennington KA. Leptin and the placental response to maternal food restriction during early pregnancy in mice. Biol Reprod 2012; 87:120. [PMID: 22993381 DOI: 10.1095/biolreprod.112.103218] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Several studies have demonstrated that maternal undernutrition or overnutrition during pregnancy can have negative consequences for the health of children born to these pregnancies, but the physiological mechanisms by which this occurs are not completely understood. During periods of food restriction, concentrations of leptin decline, whereas leptin is elevated in obesity, suggesting that it may play a role in the response to altered nutrition during pregnancy. This study compares placental development and global placental gene expression profiles at Day 11.5 in pregnant control mice, mice that were undernourished, and mice that were undernourished but given leptin. Placentas from mothers exposed to food restriction preserved the placental labyrinth zone at the expense of the junctional zone, an effect abrogated in the restricted plus leptin group, which had a significant decrease in the labyrinth zone area compared with controls. Similarly, there were more significant differences in gene expression between placentas from control and restricted plus leptin mothers (1128 differentially expressed genes) than between placentas of control and restricted mothers (281 differentially expressed genes). We conclude that the presence of high concentrations of circulating leptin during food restriction disrupts the normal adaptive response of the placenta to reduced energy availability.
Collapse
Affiliation(s)
- Laura Clamon Schulz
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, Missouri, USA.
| | | | | | | |
Collapse
|
42
|
Nuño-Ayala M, Guillén N, Arnal C, Lou-Bonafonte JM, de Martino A, García-de-Jalón JA, Gascón S, Osaba L, Osada J, Navarro MA. Cystathionine β-synthase deficiency causes infertility by impairing decidualization and gene expression networks in uterus implantation sites. Physiol Genomics 2012; 44:702-16. [DOI: 10.1152/physiolgenomics.00189.2010] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Hyperhomocysteinemia has been reported in human reproduction as a risk factor for early pregnancy loss, preeclampsia, and congenital birth defects like spina bifida. Female infertility was also observed in cystathionine beta synthase-deficient mice ( Cbs-KO) as an animal model for severe hyperhomocysteinemia. The aim for the present research was to elucidate the time-point of pregnancy loss and to pinpoint gene and cellular changes involved in the underlying pathological mechanism. By mating 90-day-old wild-type and Cbs-KO female mice with their homologous male partners, we found that pregnancy loss in Cbs-KO occurred between the 8th and 12th gestation day during placenta formation. DNA microarrays were carried out on uterus from implantation and interimplantation samples obtained on day 8. The results allowed us to select genes potentially involved in embryo death; these were individually confirmed by RT-qPCR, and their expressions were also followed throughout pregnancy. We found that changes in expression of Calb1, Ttr, Expi, Inmt, Spink3, Rpgrip1, Krt15, Mt-4, Gzmc, Gzmb, Tdo2, and Afp were important for pregnancy success, since a different regulation in Cbs-KO mice was found. Also, differences in relationships among selected genes were observed, indicating a dysregulation of these genes in Cbs-KO females. In conclusion, our data provide more information on the gene expression cascade and its timely regulated process required for a successful pregnancy. In addition, we unveil new potential avenues to explore further investigations in pregnancy loss.
Collapse
Affiliation(s)
- Mario Nuño-Ayala
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
| | - Natalia Guillén
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Arnal
- Departamento de Patología Animal, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - José M. Lou-Bonafonte
- Departamento de Fisiología y Farmacología, Facultad de Ciencias de la Salud y del Deporte, Universidad de Zaragoza, Huesca, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alba de Martino
- Departamento de Patología Animal, Facultad de Veterinaria, Universidad de Zaragoza, Zaragoza, Spain
- Unidad de Anatomía Patológica, Instituto Aragonés de Ciencias de la Salud, IIS Aragón, Zaragoza, Spain
| | | | - Sonia Gascón
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Jesús Osada
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - María-Angeles Navarro
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Veterinaria, Instituto de Investigación Sanitaria de Aragón, Universidad de Zaragoza, Zaragoza, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
43
|
Magre S, Rebourcet D, Ishaq M, Wargnier R, Debard C, Meugnier E, Vidal H, Cohen-Tannoudji J, Le Magueresse-Battistoni B. Gender differences in transcriptional signature of developing rat testes and ovaries following embryonic exposure to 2,3,7,8-TCDD. PLoS One 2012; 7:e40306. [PMID: 22808131 PMCID: PMC3392256 DOI: 10.1371/journal.pone.0040306] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 06/07/2012] [Indexed: 11/25/2022] Open
Abstract
Dioxins are persistent organic pollutants interfering with endocrine systems and causing reproductive and developmental disorders. The objective of our project was to determine the impact of an in utero exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) on reproductive function of male and female offspring in the rat with a special emphasis on the immature period. We used a low dose of TCDD (unique exposure by oral gavage of 200 ng/kg at 15.5 days of gestation) in order to mirror a response to an environmental dose of TCDD not altering fertility of the progeny. We choose a global gene expression approach using Affymetrix microarray analysis, and testes of 5 days and ovaries of 14 days of age. Less than 1% of the expressed genes in gonads were altered following embryonic TCDD exposure; specifically, 113 genes in ovaries and 56 in testes with 7 genes common to both sex gonads. It included the repressor of the aryl hydrocarbon receptor (Ahrr), the chemokines Ccl5 and Cxcl4 previously shown to be regulated by dioxin in testis, Pgds2/Hpgds and 3 others uncharacterized. To validate and extend the microarray data we realized real-time PCR on gonads at various developmental periods of interest (from 3 to 25 days for ovaries, from 5 to the adult age for testes). Overall, our results evidenced that both sex gonads responded differently to TCDD exposure. For example, we observed induction of the canonic battery of TCDD-induced genes coding enzymes of the detoxifying machinery in ovaries aged of 3–14 days of age (except Cyp1a1 induced at 3–10 days) but not in testes of 5 days (except Ahrr). We also illustrated that inflammatory pathway is one pathway activated by TCDD in gonads. Finally, we identified several new genes targeted by TCDD including Fgf13 in testis and one gene, Ptgds2/Hpgds regulated in the two sex gonads.
Collapse
Affiliation(s)
- Solange Magre
- Université Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative, EAC CNRS 4413, Paris, France
| | - Diane Rebourcet
- Université Lyon 1, INSERM U1060, INRA U1235, CarMeN, Laboratoire Lyonnais de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition, Oullins, France
| | - Muhammad Ishaq
- Université Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative, EAC CNRS 4413, Paris, France
| | - Richard Wargnier
- Université Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative, EAC CNRS 4413, Paris, France
| | - Cyrille Debard
- Université Lyon 1, INSERM U1060, INRA U1235, CarMeN, Laboratoire Lyonnais de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition, Oullins, France
| | - Emmanuelle Meugnier
- Université Lyon 1, INSERM U1060, INRA U1235, CarMeN, Laboratoire Lyonnais de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition, Oullins, France
| | - Hubert Vidal
- Université Lyon 1, INSERM U1060, INRA U1235, CarMeN, Laboratoire Lyonnais de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition, Oullins, France
| | - Joëlle Cohen-Tannoudji
- Université Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative, EAC CNRS 4413, Paris, France
| | - Brigitte Le Magueresse-Battistoni
- Université Lyon 1, INSERM U1060, INRA U1235, CarMeN, Laboratoire Lyonnais de Recherche en Cardiovasculaire, Métabolisme, Diabétologie et Nutrition, Oullins, France
- * E-mail:
| |
Collapse
|
44
|
Tunster SJ, Van de Pette M, John RM. Impact of genetic background on placental glycogen storage in mice. Placenta 2011; 33:124-7. [PMID: 22153913 DOI: 10.1016/j.placenta.2011.11.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 11/03/2011] [Accepted: 11/18/2011] [Indexed: 11/27/2022]
Abstract
129 and C57BL/6 are two of the most widely used laboratory mouse strains. While it is well known that genetic modifiers between the two strains can directly influence embryonic and adult phenotypes, less is known regarding morphological differences in placental development. Here we identify differences in the junctional zone, glycogen storage and the maternal-fetal interface between these two strains and provide examples where these differences impact the phenotypic characterisation of placental mutations.
Collapse
Affiliation(s)
- S J Tunster
- Cardiff School of Biosciences, Cardiff University, Cardiff, Wales, CF10 3AX, UK.
| | | | | |
Collapse
|
45
|
Moldenhauer LM, Keenihan SN, Hayball JD, Robertson SA. GM-CSF is an essential regulator of T cell activation competence in uterine dendritic cells during early pregnancy in mice. THE JOURNAL OF IMMUNOLOGY 2010; 185:7085-96. [PMID: 20974989 DOI: 10.4049/jimmunol.1001374] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Uterine dendritic cells (DCs) are critical for activating the T cell response mediating maternal immune tolerance of the semiallogeneic fetus. GM-CSF (CSF2), a known regulator of DCs, is synthesized by uterine epithelial cells during induction of tolerance in early pregnancy. To investigate the role of GM-CSF in regulating uterine DCs and macrophages, Csf2-null mutant and wild-type mice were evaluated at estrus, and in the periconceptual and peri-implantation periods. Immunohistochemistry showed no effect of GM-CSF deficiency on numbers of uterine CD11c(+) cells and F4/80(+) macrophages at estrus or on days 0.5 and 3.5 postcoitum, but MHC class II(+) and class A scavenger receptor(+) cells were fewer. Flow cytometry revealed reduced CD80 and CD86 expression by uterine CD11c(+) cells and reduced MHC class II in both CD11c(+) and F4/80(+) cells from GM-CSF-deficient mice. CD80 and CD86 were induced in Csf2(-/-) uterine CD11c(+) cells by culture with GM-CSF. Substantially reduced ability to activate both CD4(+) and CD8(+) T cells in vivo was evident after delivery of OVA Ag by mating with Act-mOVA males or transcervical administration of OVA peptides. This study shows that GM-CSF regulates the efficiency with which uterine DCs and macrophages activate T cells, and it is essential for optimal MHC class II- and class I-mediated indirect presentation of reproductive Ags. Insufficient GM-CSF may impair generation of T cell-mediated immune tolerance at the outset of pregnancy and may contribute to the altered DC profile and dysregulated T cell tolerance evident in infertility, miscarriage, and preeclampsia.
Collapse
Affiliation(s)
- Lachlan M Moldenhauer
- Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, Australia
| | | | | | | |
Collapse
|
46
|
El-Hashash AHK, Warburton D, Kimber SJ. Genes and signals regulating murine trophoblast cell development. Mech Dev 2009; 127:1-20. [PMID: 19755154 DOI: 10.1016/j.mod.2009.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2009] [Revised: 09/05/2009] [Accepted: 09/08/2009] [Indexed: 11/25/2022]
Abstract
A fundamental step in embryonic development is cell differentiation whereby highly specialised cell types are developed from a single undifferentiated, fertilised egg. One of the earliest lineages to form in the mammalian conceptus is the trophoblast, which contributes exclusively to the extraembryonic structures that form the placenta. Trophoblast giant cells (TGCs) in the rodent placenta form the outermost layer of the extraembryonic compartment, establish direct contact with maternal cells, and produce a number of pregnancy-specific cytokine hormones. Giant cells differentiate from proliferative trophoblasts as they exit the cell cycle and enter a genome-amplifying endocycle. Normal differentiation of secondary TGCs is a critical step toward the formation of the placenta and normal embryonic development. Trophoblast development is also of particular interest to the developmental biologist and immunobiologist, as these cells constitute the immediate cellular boundary between the embryonic and maternal tissues. Abnormalities in the development of secondary TGCs results in severe malfunction of the placenta. Herein we review new information that has been accumulated recently regarding the molecular and cellular regulation of trophoblast and placenta development. In particular, we discuss the molecular aspects of murine TGC differentiation. We also focus on the role of growth and transcription factors in TGC development.
Collapse
Affiliation(s)
- Ahmed H K El-Hashash
- Developmental Biology, Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Los Angeles, CA 90027, USA
| | | | | |
Collapse
|