1
|
Liu G, Yang K, LE Y, Wei R, Hong T, Yang J. High Vitamin D Level in Female Mice Increases the Number of Live Fetuses. J Nutr Sci Vitaminol (Tokyo) 2023; 69:1-6. [PMID: 36858535 DOI: 10.3177/jnsv.69.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Little is known about the impact of high-normal range of 25-hydroxyvitamin D [25(OH)D] on reproductive function. The aim of this study was to investigate the effect of different dose vitamin D supplementation in female mice on the pregnancy outcomes. Three groups of female mice were fed with fodder containing different dose of vitamin D at both pre-gestational and gestational stages. Serum 25(OH)D and calcium concentrations were monitored. The expression levels of vitamin D receptor (VDR) mRNA and protein in placenta were determined by real-time RT-PCR and western blot. Pregnancy outcomes were evaluated and compared among the three groups. Compared with the medium and low dose groups, serum 25(OH)D concentration was significantly increased and approximated to high-normal range in the high dose group (pre-gestational: 81.3±5.75 vs 52.8±6.24 and 25.0±3.99 ng/mL; gestational: 86.8±5.99 vs 52.6±9.29 and 27.9±4.96 ng/mL, respectively; all p<0.001). Interestingly, the average number of live fetuses per litter was much larger in the high dose group than in other two groups (19.8±5.31 vs 13.8±1.30 and 12.8±3.55 respectively, both p<0.05). However, no significant differences of the expression levels of VDR mRNA and protein in placenta were identified among the three groups. Supplementation of high dose vitamin D can enhance the female mice reproductive function. Further study is warranted to explore the mechanism by which high level of 25(OH)D in female mice increases the number of fetuses.
Collapse
Affiliation(s)
- Guoqiang Liu
- Department of Endocrinology and Metabolism, Peking University Third Hospital
| | - Kun Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital
| | - Yunyi LE
- Department of Endocrinology and Metabolism, Peking University Third Hospital
| | - Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital
| | - Jin Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital
| |
Collapse
|
2
|
Upregulation of miR-181a-5p and miR-125b-2-3p in the Maternal Circulation of Fetuses with Rh-Negative Hemolytic Disease of the Fetus and Newborn Could Be Related to Dysfunction of Placental Function. DISEASE MARKERS 2022; 2022:2594091. [PMID: 36188428 PMCID: PMC9519318 DOI: 10.1155/2022/2594091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 08/21/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022]
Abstract
The transplacental transfer of maternal antibodies to the fetus is a critical mechanism for infant protection and perinatal disease. Hemolytic disease of the fetus and newborn (HDFN) is a representative fetal disease caused by transplacental transfer of maternal IgG antibodies. However, it is unclear whether placental-related miRNAs are expressed in Rh-HDFN. Through the investigation of the miR-181a-5p and miR-125b-2-3p levels in maternal plasma using qPCR, we found that both miR-181a-5p and miR-125b-2-3p were highly expressed in maternal plasma of newborns with Rh-HDFN compared with healthy controls, indicating the potential roles of these two miRNAs in Rh-HDFN. To demonstrate whether dysregulation of miR-125b-2-3p and miR-181a-5p contributes to Rh-HDFN development, we analyze the placental miRNA-/mRNA sequencing data (GSE73714) using weighted gene coexpression network analysis (WGCNA), miRNA target predictive databases, and DAVID (Database for Annotation, Visualization, and Integrated Discovery). The results showed that miR-125b-2-3p and miR-181a-5p could regulate several biological processes including cytoplasmic microtubule organization and angiogenesis. Moreover, core promoter sequence-specific DNA binding and protein binding were highly enriched molecular functions, indicating the potential roles of transcriptional regulation. Further pathway enrichment showed that miR-181a-5p and miR-125b-2-3p could regulate several biological pathways that were closely related to placental function, including the FoxO signaling pathway, focal adhesion, mTOR signaling pathway, and central carbon metabolism in cancer. In conclusion, the present results first revealed miRNA expression in the maternal circulation of newborns with Rh-HDFN, which could be caused by dysfunction of the placenta.
Collapse
|
3
|
Tsai K, Tullis B, Jensen T, Graff T, Reynolds P, Arroyo J. Differential expression of mTOR related molecules in the placenta from gestational diabetes mellitus (GDM), intrauterine growth restriction (IUGR) and preeclampsia patients. Reprod Biol 2021; 21:100503. [PMID: 33826986 DOI: 10.1016/j.repbio.2021.100503] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/16/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023]
Abstract
The mechanistic target of rapamycin (mTOR) pathway is involved in the function and growth of the placenta during pregnancy. The mTOR pathway responds to nutrient availability and growth factors that regulate protein expression and cell growth. Disrupted mTOR signaling is associated with the development of several obstetric complications. The purpose of this study was to identify the differential placental expression of various mTOR-associated proteins in the placenta during normal gestation (Control), gestational diabetes mellitus (GDM), intrauterine growth restriction (IUGR) and preeclampsia (PE). Immunohistochemistry localized activated proteins (phospho; p) mTOR, pp70, p4EBP1, pAKT and pERK. Real-time PCR array was performed to show differing placental expression of additional mTOR-associated genes. Western blot was performed for pAMPK protein. We observed: 1) increased pmTOR during GDM and decreased pmTOR during IUGR and PE, 2) increased pp70 during IUGR and decreased pp70 during GDM and PE, 3) increased p4EBP1 during GDM, IUGR, and PE, 4) increased pAKT during GDM, 5) increased pERK during IUGR, 6) differential placental expression of mTOR pathway associated genes and increased pAMPK during GDM and PE. We conclude that regulation of the mTOR pathway is uniquely involved in the development of these obstetric complications. Insights into this pathway may provide avenues that if modify may help alleviate these diseases.
Collapse
Affiliation(s)
- Kary Tsai
- Lung and Placenta Research Laboratory, Brigham Young University, Department of Physiology and Developmental Biology, Provo, UT, USA
| | - Benton Tullis
- Lung and Placenta Research Laboratory, Brigham Young University, Department of Physiology and Developmental Biology, Provo, UT, USA
| | - Tyler Jensen
- Lung and Placenta Research Laboratory, Brigham Young University, Department of Physiology and Developmental Biology, Provo, UT, USA
| | - Taylor Graff
- Lung and Placenta Research Laboratory, Brigham Young University, Department of Physiology and Developmental Biology, Provo, UT, USA
| | - Paul Reynolds
- Lung and Placenta Research Laboratory, Brigham Young University, Department of Physiology and Developmental Biology, Provo, UT, USA
| | - Juan Arroyo
- Lung and Placenta Research Laboratory, Brigham Young University, Department of Physiology and Developmental Biology, Provo, UT, USA.
| |
Collapse
|
4
|
Kakadia JH, Jain BB, Biggar K, Sutherland A, Nygard K, Li C, Nathanielsz PW, Jansson T, Gupta MB. Hyperphosphorylation of fetal liver IGFBP-1 precedes slowing of fetal growth in nutrient-restricted baboons and may be a mechanism underlying IUGR. Am J Physiol Endocrinol Metab 2020; 319:E614-E628. [PMID: 32744097 PMCID: PMC7642856 DOI: 10.1152/ajpendo.00220.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In cultured fetal liver cells, insulin-like growth factor (IGF) binding protein (IGFBP)-1 hyperphosphorylation in response to hypoxia and amino acid deprivation is mediated by inhibition of mechanistic target of rapamycin (mTOR) and activation of amino acid response (AAR) signaling and casein kinase (CK)2. We hypothesized that fetal liver mTOR inhibition, activation of AAR and CK2, and IGFBP-1 hyperphosphorylation occur before development of intrauterine growth restriction (IUGR). Pregnant baboons were fed a control (C) or a maternal nutrient restriction (MNR; 70% calories of control) diet starting at gestational day (GD) 30 (term GD 185). Umbilical blood and fetal liver tissue were obtained at GD 120 (C, n = 7; MNR, n = 10) and 165 (C, n = 7; MNR, n = 8). Fetal weights were unchanged at GD 120 but decreased at GD 165 in the MNR group (-13%, P = 0.03). IGFBP-1 phosphorylation, as determined by parallel reaction monitoring mass spectrometry (PRM-MS), immunohistochemistry, and/or Western blot, was enhanced in MNR fetal liver and umbilical plasma at GD 120 and 165. IGF-I receptor autophosphorylationTyr1135 (-64%, P = 0.05) was reduced in MNR fetal liver at GD 120. Furthermore, fetal liver CK2 (α/α'/β) expression, CK2β colocalization, proximity with IGFBP-1, and CK2 autophosphorylationTyr182 were greater at GD 120 and 165 in MNR vs. C. Additionally, mTOR complex (mTORC)1 (p-P70S6KThr389, -52%, P = 0.05) and mTORC2 (p-AktSer473, -56%, P < 0.001) activity were decreased and AAR was activated (p-GCN2Thr898, +117%, P = 0.02; p-eIF2αSer51, +294%, P = 0.002; p-ERKThr202, +111%, P = 0.03) in MNR liver at GD 120. Our data suggest that fetal liver IGFBP-1 hyperphosphorylation, mediated by mTOR inhibition and both AAR and CK2 activation, is a key link between restricted nutrient and oxygen availability and the development of IUGR.
Collapse
Affiliation(s)
- Jenica H Kakadia
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Bhawani B Jain
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Kyle Biggar
- Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Austen Sutherland
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Karen Nygard
- Biotron Integrated Microscopy Facility, University of Western Ontario, London, Ontario, Canada
| | - Cun Li
- University of Wyoming, Laramie, Wyoming
- Southwest National Primate Research Center, San Antonio, Texas
| | - Peter W Nathanielsz
- University of Wyoming, Laramie, Wyoming
- Southwest National Primate Research Center, San Antonio, Texas
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Madhulika B Gupta
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
| |
Collapse
|
5
|
Maternal stress in relation to sex-specific expression of placental genes involved in nutrient transport, oxygen tension, immune response, and the glucocorticoid barrier. Placenta 2020; 96:19-26. [PMID: 32421529 DOI: 10.1016/j.placenta.2020.05.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/20/2020] [Accepted: 05/07/2020] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Murine models provide evidence that maternal stress during pregnancy can influence placenta morphology and function, including altered expression of genes involved in the maintenance and progression of pregnancy and fetal development. Corresponding research evaluating the impact of maternal stress on placental gene expression in humans is limited. We examined maternal stress in relation to placental expression of 17 candidate genes in a community-based sample. METHODS Participants included 60 mother-newborn pairs enrolled in the PRogramming of Intergenerational Stress Mechanisms pregnancy cohort based at the Mount Sinai Hospital in New York City. Placentas were collected immediately following delivery and gene expression was measured using a qPCR-based platform. Maternal experiences of traumatic and non-traumatic stress were measured using the Life Stressor Checklist-Revised (LSC-R) administered during a mid-pregnancy interview. We used multivariable linear regression to examine associations between LSC-R scores and expression of each gene in separate models in the sample overall and stratified by fetal sex. RESULTS Higher maternal stress was associated with significantly increased placental expression of the nutrient sensor gene OGT, the glucose transporter gene GLUT1, and the hypoxia sensor gene HIF3A. In models stratified by fetal sex, significant associations remained only among males. DISCUSSION This study represents one of the most comprehensive examinations of maternal lifetime traumatic and non-traumatic stress in relation to placental gene expression in human tissue. Our findings support that maternal stress may alter sex-specific placental expression of genes involved in critical developmental processes.
Collapse
|
6
|
Hart B, Morgan E, Alejandro EU. Nutrient sensor signaling pathways and cellular stress in fetal growth restriction. J Mol Endocrinol 2019; 62:R155-R165. [PMID: 30400060 PMCID: PMC6443503 DOI: 10.1530/jme-18-0059] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 10/09/2018] [Indexed: 12/24/2022]
Abstract
Fetal growth restriction is one of the most common obstetrical complications resulting in significant perinatal morbidity and mortality. The most frequent etiology of human singleton fetal growth restriction is placental insufficiency, which occurs secondary to reduced utero-placental perfusion, abnormal placentation, impaired trophoblast invasion and spiral artery remodeling, resulting in altered nutrient and oxygen transport. Two nutrient-sensing proteins involved in placental development and glucose and amino acid transport are mechanistic target of rapamycin (mTOR) and O-linked N-acetylglucosamine transferase (OGT), which are both regulated by availability of oxygen. Impairment in either of these pathways is associated with fetal growth restriction and accompanied by cellular stress in the forms of hypoxia, oxidative and endoplasmic reticulum (ER) stress, metabolic dysfunction and nutrient starvation in the placenta. Recent evidence has emerged regarding the potential impact of nutrient sensors on fetal stress response, which occurs in a sexual dysmorphic manner, indicating a potential element of genetic gender susceptibility to fetal growth restriction. In this mini review, we focus on the known role of mTOR and OGT in placental development, nutrient regulation and response to cellular stress in human fetal growth restriction with supporting evidence from rodent models.
Collapse
Affiliation(s)
- Bethany Hart
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Elizabeth Morgan
- Division of Maternal-Fetal Medicine, Department of Obstetrics, Gynecology, and Women's Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Emilyn U Alejandro
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
7
|
Chudasama D, Bo V, Hall M, Anikin V, Jeyaneethi J, Gregory J, Pados G, Tucker A, Harvey A, Pink R, Karteris E. Identification of cancer biomarkers of prognostic value using specific gene regulatory networks (GRN): a novel role of RAD51AP1 for ovarian and lung cancers. Carcinogenesis 2018; 39:407-417. [PMID: 29126163 PMCID: PMC5862298 DOI: 10.1093/carcin/bgx122] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022] Open
Abstract
To date, microarray analyses have led to the discovery of numerous individual ‘molecular signatures’ associated with specific cancers. However, there are serious limitations for the adoption of these multi-gene signatures in the clinical environment for diagnostic or prognostic testing as studies with more power need to be carried out. This may involve larger richer cohorts and more advanced analyses. In this study, we conduct analyses—based on gene regulatory network—to reveal distinct and common biomarkers across cancer types. Using microarray data of triple-negative and medullary breast, ovarian and lung cancers applied to a combination of glasso and Bayesian networks (BNs), we derived a unique network-containing genes that are uniquely involved: small proline-rich protein 1A (SPRR1A), follistatin like 1 (FSTL1), collagen type XII alpha 1 (COL12A1) and RAD51 associated protein 1 (RAD51AP1). RAD51AP1 and FSTL1 are significantly overexpressed in ovarian cancer patients but only RAD51AP1 is upregulated in lung cancer patients compared with healthy controls. The upregulation of RAD51AP1 was mirrored in the bloods of both ovarian and lung cancer patients, and Kaplan–Meier (KM) plots predicted poorer overall survival (OS) in patients with high expression of RAD51AP1. Suppression of RAD51AP1 by RNA interference reduced cell proliferation in vitro in ovarian (SKOV3) and lung (A549) cancer cells. This effect appears to be modulated by a decrease in the expression of mTOR-related genes and pro-metastatic candidate genes. Our data describe how an initial in silico approach can generate novel biomarkers that could potentially support current clinical practice and improve long-term outcomes.
Collapse
Affiliation(s)
- Dimple Chudasama
- Institute for Environment, Health and Societies, Brunel University London, Uxbridge, UK
| | - Valeria Bo
- Department of Computer Science, Brunel University London, Uxbridge, UK.,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | | | - Vladimir Anikin
- Department of Cardiothoracic Surgery, Harefield Hospital, Royal Brompton and Harefield Trust, Harefield, UK
| | - Jeyarooban Jeyaneethi
- Institute for Environment, Health and Societies, Brunel University London, Uxbridge, UK
| | - Jane Gregory
- Department of Cardiothoracic Surgery, Harefield Hospital, Royal Brompton and Harefield Trust, Harefield, UK
| | - George Pados
- University of Thessaloniki Medical School, Thessaloniki, Greece
| | - Allan Tucker
- Department of Computer Science, Brunel University London, Uxbridge, UK
| | - Amanda Harvey
- Institute for Environment, Health and Societies, Brunel University London, Uxbridge, UK
| | - Ryan Pink
- Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, UK
| | - Emmanouil Karteris
- Institute for Environment, Health and Societies, Brunel University London, Uxbridge, UK
| |
Collapse
|
8
|
MANKARIOUS AMANDA, DAVE FORAM, PADOS GEORGE, TSOLAKIDIS DIMITRIS, GIDRON YORI, PANG YEFEI, THOMAS PETER, HALL MARCIA, KARTERIS EMMANOUIL. The pro-social neurohormone oxytocin reverses the actions of the stress hormone cortisol in human ovarian carcinoma cells in vitro. Int J Oncol 2016; 48:1805-14. [PMID: 26935408 PMCID: PMC4809651 DOI: 10.3892/ijo.2016.3410] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 12/18/2015] [Indexed: 12/16/2022] Open
Abstract
The journey patients with ovarian cancer travel from non-specific symptoms causing delayed diagnosis through surgery and chemotherapy, culminating in a 5-year survival rate of 43%, must have a profound and detrimental psychological impact on patients. Emerging studies link higher levels of oxytocin (OT) and increased social support, an independent prognostic factor in cancer, with a moderating effect on stress. In contrast, there is a known association of tumour cell proliferation with elevated cortisol (stress hormone) levels. We hypothesise therefore that there is cross-talk between cortisol and oxytocin at a molecular level. Three ovarian cancer cell lines, used as in vitro models, were treated with cortisol at concentrations mimicking physiological stress in vivo in the presence or absence of OT. OT reduced cell proliferation and migration, induced apoptosis and autophagy for all three cell lines, partially reversing the effects of cortisol. Quantitative RT-PCR of tissue taken from ovarian cancer patients revealed that the glucocorticoid receptor (splice variant GR-P) and OT receptor (OTR) were significantly upregulated compared to controls. Tissue microarray revealed that the expression of GRα was lower in the ovarian cancer samples compared to normal tissue. OT is also shown to drive alternative splicing of the GR gene and cortisol-induced OTR expression. OT was able to transactivate GR in the presence of cortisol, thus providing further evidence of cross-talk in vitro. These data provide explanations for why social support might help distressed ovarian cancer patients and help define novel hypotheses regarding potential therapeutic interventions in socially isolated patients.
Collapse
Affiliation(s)
- AMANDA MANKARIOUS
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
| | - FORAM DAVE
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
| | - GEORGE PADOS
- University of Thessaloniki Medical School, Thessaloniki, Greece
| | | | - YORI GIDRON
- Free University of Brussels (VUB), Brussels, Belgium
| | - YEFEI PANG
- University of Texas at Austin, Marine Science Institute, Port Aransas, TX 78373, USA
| | - PETER THOMAS
- University of Texas at Austin, Marine Science Institute, Port Aransas, TX 78373, USA
| | - MARCIA HALL
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
- Mount Vernon Cancer Centre, Northwood HA6 2RN, UK
| | - EMMANOUIL KARTERIS
- Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
- Correspondence to: Dr Emmanouil Karteris, Division of Biosciences, Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge UB8 3PH, UK, E-mail:
| |
Collapse
|
9
|
He B, Zhang N, Zhao R. Dexamethasone Downregulates SLC7A5 Expression and Promotes Cell Cycle Arrest, Autophagy and Apoptosis in BeWo Cells. J Cell Physiol 2016; 231:233-42. [PMID: 26094588 DOI: 10.1002/jcp.25076] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/08/2015] [Indexed: 12/14/2022]
Abstract
Synthetic glucocorticoids (GCs) such as dexamethasone (Dex) are widely given to pregnant women to induce maturation and improve viability of preterm infants. Despite the beneficial effects, synthetic GCs have adverse effects on placental growth and nutrient transport system. However, the molecular mechanisms involved in these events remain unknown. Here we use a human placental choriocarcinoma cell line (BeWo) as model to explore the pathway linking amino acids transport with cell viability under Dex challenge. BeWo cells treated with Dex (100 nM) for 24 h demonstrated G1/S cell cycle arrest together with enhanced autophagy and apoptosis. Concurrently, the amino acid carrier SLC7A5 was down-regulated in association with impaired cellular amino acids uptake and inhibition of mammalian target of rapamycin (mTOR) signaling. Similar cellular responses were observed in BeWo cells treated with BCH, a classical System L inhibitor which inactivates SLC7A5. The glucocorticoid receptor (GR) antagonist RU486 was able to diminish Dex-induced translocation of GR into nucleus and to abolish these effects. Furthermore, Dex treatment significantly promoted the binding of GR to the proximal promoter sequence of SLC7A5 gene. Taken together, our results show that Dex downregulates SLC7A5 expression via GR-mediated transrepression. The impaired amino acids uptake leads to inhibition of mTOR signaling which in turn causes inhibited proliferation and enhanced autophagy and apoptosis in BeWo cells. These findings indicate that SLC7A5 mediates the effect of Dex on cell viability, thus providing a novel molecular target for the prevention and treatment of Dex-induced cell cycle arrest and apoptosis in placental cells.
Collapse
Affiliation(s)
- Bin He
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| | - Nana Zhang
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| | - Ruqian Zhao
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
10
|
Sati L, Soygur B, Celik-Ozenci C. Expression of Mammalian Target of Rapamycin and Downstream Targets in Normal and Gestational Diabetic Human Term Placenta. Reprod Sci 2015; 23:324-32. [DOI: 10.1177/1933719115602765] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Leyla Sati
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Bikem Soygur
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - Ciler Celik-Ozenci
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| |
Collapse
|
11
|
Mina TH, Räikkönen K, Riley SC, Norman JE, Reynolds RM. Maternal distress associates with placental genes regulating fetal glucocorticoid exposure and IGF2: Role of obesity and sex. Psychoneuroendocrinology 2015; 59:112-22. [PMID: 26056743 DOI: 10.1016/j.psyneuen.2015.05.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 04/16/2015] [Accepted: 05/08/2015] [Indexed: 01/01/2023]
Abstract
Maternal emotional distress symptoms, including life satisfaction, anxiety and depressed mood, are worse in Severely Obese (SO) than lean pregnancy and may alter placental genes regulating fetal glucocorticoid exposure and placental growth. We hypothesised that the associations between increased maternal distress symptoms and changes in placental gene expression including IGF2 and genes regulating fetal glucocorticoid exposure are more pronounced in SO pregnancy. We also considered whether there were sex-specific effects. Placental mRNA levels of 11β-HSDs, NR3C1-α, NR3C2, ABC transporters, mTOR and the IGF2 family were measured in term placental samples from 43 lean (BMI≤25kg/m(2)) and 50 SO (BMI≥40kg/m(2)) women, in whom distress symptoms were prospectively evaluated during pregnancy. The mRNA levels of genes with a similar role in regulating fetal glucocorticoid exposure were strongly inter-correlated. Increased maternal distress symptoms associated with increased NR3C2 and IGF2 isoform 1(IGF2-1) in both lean and SO group (p≤0.05). Increased distress was associated with higher ABCB1 and ABCG2 mRNA levels in SO but lower ABCB1 and higher 11β-HSD1 mRNA levels in lean (p≤0.05) suggesting a protective adaptive response in SO placentas. Increased maternal distress associated with reduced mRNA levels of ABCB1, ABCG2, 11β-HSD2, NR3C1-α and IGF2-1 in placentas of female but not male offspring. The observed sex differences in placental responses suggest greater vulnerability of female fetuses to maternal distress with potentially greater fetal glucocorticoid exposure and excess IGF2. Further studies are needed to replicate these findings and to test whether this translates to potentially greater negative outcomes of maternal distress in female offspring in early childhood.
Collapse
Affiliation(s)
- Theresia H Mina
- University BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK; Tommy's Centre for Maternal and Fetal Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, UK
| | - Katri Räikkönen
- Institute of Behavioral Sciences, University of Helsinki, 00014 Helsinki, Finland
| | - Simon C Riley
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, UK; Tommy's Centre for Maternal and Fetal Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, UK
| | - Jane E Norman
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, UK; Tommy's Centre for Maternal and Fetal Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, UK
| | - Rebecca M Reynolds
- University BHF Centre for Cardiovascular Sciences, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK; Tommy's Centre for Maternal and Fetal Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, UK.
| |
Collapse
|
12
|
Davies J, Chen J, Pink R, Carter D, Saunders N, Sotiriadis G, Bai B, Pan Y, Howlett D, Payne A, Randeva H, Karteris E. Orexin receptors exert a neuroprotective effect in Alzheimer's disease (AD) via heterodimerization with GPR103. Sci Rep 2015; 5:12584. [PMID: 26223541 PMCID: PMC4519789 DOI: 10.1038/srep12584] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 05/29/2015] [Indexed: 12/22/2022] Open
Abstract
Orexins are neuropeptides that regulate the sleep-wake cycle and feeding behaviour. QRFP is a newly discovered neuropeptide which exerts similar orexigenic activity, thus playing an important role in energy homeostasis and regulation of appetite. The exact expression and signalling characteristics and physiological actions of QRFP and its receptor GPR103 are poorly understood. Alzheimer's disease (AD) patients experience increased nocturnal activity, excessive daytime sleepiness, and weight loss. We hypothesised therefore that orexins and QRFP might be implicated in the pathophysiology of AD. We report that the down-regulation of hippocampal orexin receptors (OXRs) and GPR103 particularly in the cornu ammonis (CA) subfield from AD patients suffering from early onset familial AD (EOFAD) and late onset familial AD (LOAD). Using an in vitro model we demonstrate that this downregulation is due to to Aβ-plaque formation and tau hyper-phosphorylation. Transcriptomics revealed a neuroprotective role for both orexins and QRFP. Finally we provide conclusive evidence using BRET and FRET that OXRs and GPR103 form functional hetero-dimers to exert their effects involving activation of ERK1/2. Pharmacological intervention directed at the orexigenic system may prove to be an attractive avenue towards the discovery of novel therapeutics for diseases such as AD and improving neuroprotective signalling pathways.
Collapse
Affiliation(s)
- Julie Davies
- Biosciences, College of Health and Life Sciences, Brunel University, Uxbridge, UB8 3PH, UK
| | - Jing Chen
- Division of Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, P.R. China
| | - Ryan Pink
- Department of Biological and Medical Sciences, Oxford Brookes University, UK
| | - David Carter
- Department of Biological and Medical Sciences, Oxford Brookes University, UK
| | - Nigel Saunders
- Centre for Systems and Synthetic Biology, Brunel University, Uxbridge UB83PH, UK
| | - Georgios Sotiriadis
- Biosciences, College of Health and Life Sciences, Brunel University, Uxbridge, UB8 3PH, UK
| | - Bo Bai
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, P.R. China
| | - Yanyou Pan
- Neurobiology Institute, Jining Medical University, Jining, Shandong, 272067, P.R. China
| | - David Howlett
- Wolfson Centre for Age Related Diseases, King’s College London, London, SE11UL, UK
| | - Annette Payne
- Department of Computer Science, College of Engineering, Design and Physical Sciences, Brunel University, Uxbridge UB8 3PH, UK
| | - Harpal Randeva
- Division of Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham, B4 7ET, UK
| | - Emmanouil Karteris
- Biosciences, College of Health and Life Sciences, Brunel University, Uxbridge, UB8 3PH, UK
| |
Collapse
|
13
|
Davies J, Zachariades E, Rogers-Broadway KR, Karteris E. Elucidating the role of DEPTOR in Alzheimer's disease. Int J Mol Med 2014; 34:1195-200. [PMID: 25119265 PMCID: PMC4199409 DOI: 10.3892/ijmm.2014.1895] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 07/29/2014] [Indexed: 01/22/2023] Open
Abstract
The mammalian or mechanistic target of rapamycin (mTOR) is a Ser/Thr protein kinase that, in response to nutrient stimulation, regulates cellular growth, proliferation, survival, protein synthesis and gene transcription. It has also been implicated in Alzheimer’s disease (AD) with neuronal cells and hippocampal slices of AD transgenic mice experiencing dysregulated mTOR and synaptic plasticity in response to treatment with the toxic amyloid β (Aβ1–42) peptide, which has been implicated in AD. DEP domain-containing mTOR-interacting protein (DEPTOR) is a protein which can bind to mTOR and cause its inhibition, and functions as a regulatory protein of mTOR to control its activity. The inhibition of mTOR has been shown to have a neuroprotective effect; in an animal model, it was shown to protect against Aβ-induced neurotoxicity. In the present study, to investigate to role of DEPTOR in a model of AD, we neuronally differentiated the SH-SY5Y cell line and examined the effects of treatment with an Aβ42 peptide, thus mimicking plaque formation. This resulted in a significant increase in mTOR and a significant decrease in DEPTOR expression compared to the unstimulated controls. Moreover, to the best of our knowledge, we demonstrate for the first time a reduction in the protein level of DEPTOR in the precentral gyrus, postcentral gyrus and occipital lobe of a brain with AD compared to a normal control, as well as a significant reduction in DEPTOR expression in samples from late-onset AD (LOAD) compared to early-onset familial AD (EOFAD). The reduction in DEPTOR expression in cases of AD compared to healthy controls can lead to an augmentation of mTOR signalling, leading to Aβ accumulation, which in turn leads to a further reduction in DEPTOR expression. This results in the accumulation of amyloid plaque, shifting the balance from neuroprotection to neurodegeneration.
Collapse
Affiliation(s)
- Julie Davies
- Department of Biosciences, School of Health Sciences and Social Care, Brunel University, Uxbridge, UB8 3PH, UK
| | - Elena Zachariades
- Department of Biosciences, School of Health Sciences and Social Care, Brunel University, Uxbridge, UB8 3PH, UK
| | - Karly-Rai Rogers-Broadway
- Department of Biosciences, School of Health Sciences and Social Care, Brunel University, Uxbridge, UB8 3PH, UK
| | - Emmanouil Karteris
- Department of Biosciences, School of Health Sciences and Social Care, Brunel University, Uxbridge, UB8 3PH, UK
| |
Collapse
|
14
|
Differential expression of placental glucocorticoid receptors and growth arrest-specific transcript 5 in term and preterm pregnancies: evidence for involvement of maternal stress. Obstet Gynecol Int 2014; 2014:239278. [PMID: 24899900 PMCID: PMC4037583 DOI: 10.1155/2014/239278] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 01/18/2014] [Accepted: 03/12/2014] [Indexed: 02/08/2023] Open
Abstract
Pregnancy-specific stress predicts birth outcomes. We hypothesized that there is a maternal stress-GR interaction that can influence fetal birth weight. This study examined the relationship between mothers' stress and attitude towards their pregnancies, placental glucocorticoid receptors (GRs) and growth arrest-specific transcript 5 (GAS5) expression, and the status of GR polymorphism, with their infants' birth weights. GAS5 and GRα were the predominant transcripts in both term and preterm placentas, with GAS5 being primarily localized in the syncytiotrophoblasts. In an attempt to mimic moderate and high stress environment in vitro, BeWo and JEG-3 cytotrophoblast cell lines were treated with 10 nM–1000 nM cortisol. Only expression of GAS5 was significantly upregulated by cortisol in all treatments compared with basal levels, but none of the GRs changed expression significantly. In an attempt to assess a stress versus gene interaction, we studied four GR polymorphisms. In the homozygous group for Tth111I polymorphism, mothers with negative attitudes towards the pregnancy gave birth to infants with significantly lower birth weights compared to women with positive/neutral attitudes. None of the GR splice variants were associated with maternal stress. However, placental GAS5 levels were inversely correlated with maternal stress. This study points towards a potential gene-environment interaction that could be of predictive value for fetal weight.
Collapse
|
15
|
Hogg K, Robinson WP, Beristain AG. Activation of endocrine-related gene expression in placental choriocarcinoma cell lines following DNA methylation knock-down. Mol Hum Reprod 2014; 20:677-89. [PMID: 24623739 DOI: 10.1093/molehr/gau020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Increasingly, placental DNA methylation is assessed as a factor in pregnancy-related complications, yet the transcriptional impact of such findings is not always clear. Using a proliferative in vitro placental model, the effect of DNA methylation loss on gene activation was evaluated at a number of genes selected for being differentially methylated in pre-eclampsia-associated placentae in vivo. We aimed to determine whether reduced DNA methylation at specific loci was associated with transcriptional changes at the corresponding gene, thus providing mechanistic underpinnings for previous clinical findings and to assess the degree of transcriptional response amongst our candidate genes. BeWo and JEG3 choriocarcinoma cells were exposed to 1 μM 5-Aza-2'-deoxycytidine (5-Aza-CdR) or vehicle control for 48 h, and re-plated and cultured for a further 72 h in normal media before cells were harvested for RNA and DNA. Bisulphite pyrosequencing confirmed that DNA methylation was reduced by ∼30-50% points at the selected loci studied in both cell lines. Gene activation, measured by qRT-PCR, was highly variable and transcript specific, indicating differential sensitivity to DNA methylation. Most notably, loss of DNA methylation at the leptin (LEP) promoter corresponded to a 200-fold and 40-fold increase in LEP expression in BeWo and JEG3 cells, respectively (P < 0.01). Transcripts of steroidogenic pathway enzymes CYP11A1 and HSD3B1 were up-regulated ∼40-fold in response to 5-Aza-CdR exposure in BeWo cells (P < 0.01). Other transcripts, including aromatase (CYP19), HSD11B2, inhibin (INHBA) and glucocorticoid receptor (NR3C1) were more moderately, although significantly, affected by loss of associated DNA methylation. These data present a mixed effect of DNA methylation changes at selected loci supporting cautionary interpretation of DNA methylation results in the absence of functional data.
Collapse
Affiliation(s)
- K Hogg
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada Child & Family Research Institute, Vancouver, BC, Canada
| | - W P Robinson
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada Child & Family Research Institute, Vancouver, BC, Canada
| | - A G Beristain
- Child & Family Research Institute, Vancouver, BC, Canada Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Armstrong DA, Lesseur C, Conradt E, Lester BM, Marsit CJ. Global and gene-specific DNA methylation across multiple tissues in early infancy: implications for children's health research. FASEB J 2014; 28:2088-97. [PMID: 24478308 DOI: 10.1096/fj.13-238402] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
An increasing number of population studies are assessing epigenetic variation in relation to early-life outcomes in tissues accessible to epidemiologic researchers. Epigenetic mechanisms are highly tissue specific, however, and it is unclear whether the variation observed in one of the tissue types is representative of other sources or whether the variation in DNA methylation is distinct, reflecting potential functional differences across tissues. To assess relations between DNA methylation in various samples from newborns and children in early infancy, we measured promoter or gene-body DNA methylation in matched term placenta, cord blood, and 3-6 mo saliva samples from 27 unrelated infants enrolled in the Rhode Island Child Health Study. We investigated 7 gene loci (KLF15, NR3C1, LEP, DEPTOR, DDIT4, HSD11B2, and CEBPB) and global methylation, using repetitive region LINE-1 and ALUYb8 sequences. We observed a great degree of interlocus, intertissue, and interindividual epigenetic variation in most of the analyzed loci. In correlation analyses, only cord blood NR3C1 promoter methylation correlated negatively with methylation in saliva. We conclude that placenta, cord blood, and saliva cannot be used as a substitute for one another to evaluate DNA methylation at these loci during infancy. Each tissue has a unique epigenetic signature that likely reflects their differential functions. Future studies should consider the uniqueness of these features, to improve epigenetic biomarker discovery and translation.
Collapse
Affiliation(s)
- David A Armstrong
- 2Department of Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, 7650 Remsen, Hanover, NH 03755, USA.
| | | | | | | | | |
Collapse
|
17
|
Foster HA, Davies J, Pink RC, Turkcigdem S, Goumenou A, Carter DR, Saunders NJ, Thomas P, Karteris E. The human myometrium differentially expresses mTOR signalling components before and during pregnancy: evidence for regulation by progesterone. J Steroid Biochem Mol Biol 2014; 139:166-72. [PMID: 23541542 PMCID: PMC3855612 DOI: 10.1016/j.jsbmb.2013.02.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 02/07/2013] [Accepted: 02/24/2013] [Indexed: 10/27/2022]
Abstract
Emerging studies implicate the signalling of the mammalian target of rapamycin (mTOR) in a number of reproductive functions. To this date, there are no data regarding the expression of mTOR signalling components in the human myometrium during pregnancy. We hypothesized that mTOR-related genes might be differentially expressed in term or preterm labour as well as in labour or non-labour myometria during pregnancy. Using quantitative RT-PCR we demonstrate for first time that there is a significant downregulation of mTOR, DEPTOR, and Raptor in preterm labouring myometria when compared to non-pregnant tissues taken from the same area (lower segment). We used an immortalized myometrial cell line (ULTR) as an in vitro model to dissect further mTOR signalling. In ULTR cells DEPTOR and Rictor had a cytoplasmic distribution, whereas mTOR and Raptor were detected in the cytoplasm and the nucleus, indicative of mTORC1 shuttling. Treatment with inflammatory cytokines caused only minor changes in gene expression of these components, whereas progesterone caused significant down-regulation. We performed a non-biased gene expression analysis of ULTR cells using Nimblegen human gene expression microarray (n=3), and selected genes were validated by quantitative RT-PCR in progesterone treated myometrial cells. Progesterone significantly down-regulated key components of the mTOR pathway. We conclude that the human myometrium differentially expresses mTOR signalling components and they can be regulated by progesterone. This article is part of a Special Issue entitled 'Pregnancy and Steroids'.
Collapse
Affiliation(s)
- Helen A Foster
- Biosciences, Centre for Cell and Chromosome Biology, Brunel University, Uxbridge UB8 3PH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Mina TH, Reynolds RM. Mechanisms linking in utero stress to altered offspring behaviour. Curr Top Behav Neurosci 2014; 18:93-122. [PMID: 24577734 DOI: 10.1007/7854_2014_291] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Development in utero is recognised as a determinant of health in later life, a concept known as early life 'programming'. Several studies in humans have now shown a link between in utero stressors of maternal stress, anxiety and depression and adverse behavioural outcomes for the offspring including poorer cognitive function and behavioural and emotional problems. These behaviours are observed from the very early neonatal period and appear to persist through to adulthood. Underlying mechanisms are not known but overexposure of the developing foetus to glucocorticoids has been proposed. Dysregulation of the maternal and offspring hypothalamic-pituitary-adrenal (HPA) axis has been proposed as a mechanism linking in utero stress with offspring behavioural outcomes. Studies suggest that altered circulating levels of maternal cortisol during pregnancy and/or changes in placental gene expression or methylation, which result in increased glucocorticoid transfer to the developing foetus, are linked to changes in offspring behaviour and in activity of the offspring HPA axis. Further understanding of the underlying pathways and identification of any gestation of vulnerability are needed to help design interventions to reduce in utero stress and improve behavioural outcomes in the offspring.
Collapse
Affiliation(s)
- Theresia H Mina
- Endocrinology Unit, Queen's Medical Research Institute, University/British Heart Foundation Centre for Cardiovascular Science, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | | |
Collapse
|
19
|
Braun T, Challis JR, Newnham JP, Sloboda DM. Early-life glucocorticoid exposure: the hypothalamic-pituitary-adrenal axis, placental function, and long-term disease risk. Endocr Rev 2013; 34:885-916. [PMID: 23970762 DOI: 10.1210/er.2013-1012] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
An adverse early-life environment is associated with long-term disease consequences. Adversity early in life is hypothesized to elicit developmental adaptations that serve to improve fetal and postnatal survival and prepare the organism for a particular range of postnatal environments. These processes, although adaptive in their nature, may later prove to be maladaptive or disadvantageous if the prenatal and postnatal environments are widely discrepant. The exposure of the fetus to elevated levels of either endogenous or synthetic glucocorticoids is one model of early-life adversity that contributes substantially to the propensity of developing disease. Moreover, early-life glucocorticoid exposure has direct clinical relevance because synthetic glucocorticoids are routinely used in the management of women at risk of early preterm birth. In this regard, reports of adverse events in human newborns have raised concerns about the safety of glucocorticoid treatment; synthetic glucocorticoids have detrimental effects on fetal growth and development, childhood cognition, and long-term behavioral outcomes. Experimental evidence supports a link between prenatal exposure to synthetic glucocorticoids and alterations in fetal development and changes in placental function, and many of these alterations appear to be permanent. Because the placenta is the conduit between the maternal and fetal environments, it is likely that placental function plays a key role in mediating effects of fetal glucocorticoid exposure on hypothalamic-pituitary-adrenal axis development and long-term disease risk. Here we review recent insights into how the placenta responds to changes in the intrauterine glucocorticoid environment and discuss possible mechanisms by which the placenta mediates fetal hypothalamic-pituitary-adrenal development, metabolism, cardiovascular function, and reproduction.
Collapse
Affiliation(s)
- Thorsten Braun
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, 1280 Main Street West, HSC 4H30A, Hamilton, Ontario, Canada L8S 4K1.
| | | | | | | |
Collapse
|
20
|
Chen B, Longtine MS, Nelson DM. Hypoxia induces autophagy in primary human trophoblasts. Endocrinology 2012; 153:4946-54. [PMID: 22878401 PMCID: PMC3512007 DOI: 10.1210/en.2012-1472] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 07/16/2012] [Indexed: 01/08/2023]
Abstract
Autophagy is a highly regulated and dynamic process that maintains cellular homeostasis and plays a prosurvival role in most cells. Although hypoxia has been shown to induce apoptosis in placental trophoblasts, the hypoxic effect on autophagy has not been studied. We hypothesized that autophagy plays a prosurvival role in the placental trophoblasts by antagonizing hypoxia-induced apoptosis. Our data show that the expression of Light chain 3-II (LC3-II), an autophagic marker and cleaved poly(ADP-ribose) polymerase, an apoptosis marker, are inversely related in cultured trophoblasts. Exposure to rapamycin or hypoxia inactivated mammalian target of rapamycin, as reflected by reduced phosphorylation of ribosomal protein S6, indicating that mammalian target of rapamycin regulates autophagy in cultured cytotrophoblasts. Bafilomycin prevented the degradation of cargo and increased LC3-II and p62 in cytotrophoblasts exposed to hypoxia, revealing enhanced autophagic flux. Importantly, bafilomycin enhanced expression of autophagy-related protein 7 (Atg7), parallel to the increased apoptosis measured by cleaved poly(ADP-ribose) polymerase. LY294002, a phosphatidylinositol 3-kinase inhibitor, increased apoptosis in the trophoblasts under hypoxia or standard conditions. Silencing of Atg7 decreased both apoptosis and LC3-II in the trophoblasts, suggesting a dual role of Atg7 in both autophagy and apoptosis. We conclude that there is a cross talk between autophagy and apoptosis in the placental trophoblasts; autophagy plays a prosurvival role and Atg7 has roles in both autophagy and apoptosis under hypoxia.
Collapse
Affiliation(s)
- Baosheng Chen
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St Louis, Missouri 63110, USA.
| | | | | |
Collapse
|
21
|
Bildirici I, Longtine MS, Chen B, Nelson DM. Survival by self-destruction: a role for autophagy in the placenta? Placenta 2012; 33:591-8. [PMID: 22652048 PMCID: PMC3389146 DOI: 10.1016/j.placenta.2012.04.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 04/17/2012] [Accepted: 04/17/2012] [Indexed: 12/20/2022]
Abstract
Autophagy is a burgeoning area of research from yeast to humans. Although previously described as a death pathway, autophagy is now considered an important survival phenomenon in response to environmental stressors to which most organs are exposed. Despite an ever expanding literature in non-placental cells, studies of autophagy in the placenta are lagging. We review the regulation of autophagy, summarize available placental studies of autophagy, and highlight potential areas for future research. We believe that such studies will yield novel insights into how placentas protect the survival of the species by "self-eating".
Collapse
Affiliation(s)
- I Bildirici
- Department of Obstetrics and Gynecology, Acibadem University School of Medicine, Istanbul, Turkey.
| | | | | | | |
Collapse
|