51
|
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
|
52
|
Melbourne A, Schabel MC, David AL, Roberts VHJ. Magnetic resonance imaging of placental intralobule structure and function in a preclinical nonhuman primate model†. Biol Reprod 2024; 110:1065-1076. [PMID: 38442734 PMCID: PMC11180614 DOI: 10.1093/biolre/ioae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/25/2024] [Accepted: 03/04/2024] [Indexed: 03/07/2024] Open
Abstract
Although the central role of adequate blood flow and oxygen delivery is known, the lack of optimized imaging modalities to study placental structure has impeded our understanding of its vascular function. Magnetic resonance imaging is increasingly being applied in this field, but gaps in knowledge remain, and further methodological developments are needed. In particular, the ability to distinguish maternal from fetal placental perfusion and the understanding of how individual placental lobules are functioning are lacking. The potential clinical benefits of developing noninvasive tools for the in vivo assessment of blood flow and oxygenation, two key determinants of placental function, are tremendous. Here, we summarize a number of structural and functional magnetic resonance imaging techniques that have been developed and applied in animal models and studies of human pregnancy over the past decade. We discuss the potential applications and limitations of these approaches. Their combination provides a novel source of contrast to allow analysis of placental structure and function at the level of the lobule. We outline the physiological mechanisms of placental T2 and T2* decay and devise a model of how tissue composition affects the observed relaxation properties. We apply this modeling to longitudinal magnetic resonance imaging data obtained from a preclinical pregnant nonhuman primate model to provide initial proof-of-concept data for this methodology, which quantifies oxygen transfer and placental structure across and between lobules. This method has the potential to improve our understanding and clinical management of placental insufficiency once validation in a larger nonhuman primate cohort is complete.
Collapse
Affiliation(s)
- Andrew Melbourne
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
- Department of Obstetrics and Maternal Fetal Medicine, Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, UK
| | - Matthias C Schabel
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, USA
- Utah Center for Advanced Imaging Research, University of Utah, Salt Lake City, Utah, USA
| | - Anna L David
- Department of Obstetrics and Maternal Fetal Medicine, Elizabeth Garrett Anderson Institute for Women’s Health, University College London, London, UK
| | - Victoria H J Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
53
|
Wilson RL, Davenport BN, Jones HN. Mid-pregnancy placental transcriptome in a model of placental insufficiency with and without novel intervention. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.05.597621. [PMID: 38895312 PMCID: PMC11185618 DOI: 10.1101/2024.06.05.597621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Fetal growth restriction (FGR) affects between 5-10% of all live births. Placental insufficiency is a leading cause of FGR, resulting in reduced nutrient and oxygen delivery to the fetus. Currently, there are no effective in utero treatment options for FGR, or placental insufficiency. We have developed a gene therapy to deliver, via a non-viral nanoparticle, human insulin-like 1 growth factor ( hIGF1 ) to the placenta as potential treatment of placenta insufficiency and FGR. Using a guinea pig maternal nutrient restriction (MNR) model of FGR, we aimed to understand the transcriptional changes within the placenta associated with placental insufficiency that occur prior to/at initiation of FGR, and the impact of short-term hIGF1 nanoparticle treatment. Using RNAsequencing, we analyzed protein coding genes of three experimental groups: Control and MNR dams receiving a sham treatment, and MNR dams receiving hIGF1 nanoparticle treatment. Pathway enrichment analysis comparing differentially expressed genelists in sham-treated MNR placentas to Control revealed upregulation of pathways associated with degradation and repair of genetic information and downregulation of pathways associated with transmembrane transport. When compared to sham-treated MNR placentas, MNR + hIGF1 placentas demonstrated changes to genelists associated with transmembrane transporter activity including ion, vitamin and solute carrier transport. Overall, this study identifies the key signaling and metabolic changes occurring in the placenta contributing to placental insufficiency prior to/at initiation of FGR, and increases our understanding of the pathways that our nanoparticle-mediated gene therapy intervention regulates. Statements and Declarations Competing Interests: Authors declare no conflicts of interest.
Collapse
|
54
|
Yaguchi C, Ueda M, Mizuno Y, Fukuchi C, Matsumoto M, Furuta-Isomura N, Itoh H. Association of Placental Pathology with Physical and Neuronal Development of Infants: A Narrative Review and Reclassification of the Literature by the Consensus Statement of the Amsterdam Placental Workshop Group. Nutrients 2024; 16:1786. [PMID: 38892717 PMCID: PMC11174896 DOI: 10.3390/nu16111786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
The placenta is the largest fetal organ, which connects the mother to the fetus and supports most aspects of organogenesis through the transport of nutrients and gases. However, further studies are needed to assess placental pathology as a reliable predictor of long-term physical growth or neural development in newborns. The Consensus Statement of the Amsterdam Placental Workshop Group (APWGCS) on the sampling and definition of placental lesions has resulted in diagnostic uniformity in describing the most common pathological lesions of the placenta and contributed to the international standardization of descriptions of placental pathology. In this narrative review, we reclassified descriptions of placental pathology from previously published papers according to the APWGCS criteria and comparatively assessed the relationship with infantile physical and/or neural development. After reclassification and reevaluation, placental pathology of maternal vascular malperfusion, one of the APWGCS criteria, emerged as a promising candidate as a universal predictor of negative infantile neurodevelopmental outcomes, not only in term and preterm deliveries but also in high-risk groups of very low birthweight newborns. However, there are few studies that examined placental pathology according to the full categories of APWGCS and also included low-risk general infants. It is necessary to incorporate the assessment of placental pathology utilizing APWGCS in the design of future birth cohort studies as well as in follow-up investigations of high-risk infants.
Collapse
|
55
|
Cristodoro M, Messa M, Tossetta G, Marzioni D, Dell’Avanzo M, Inversetti A, Di Simone N. First Trimester Placental Biomarkers for Pregnancy Outcomes. Int J Mol Sci 2024; 25:6136. [PMID: 38892323 PMCID: PMC11172712 DOI: 10.3390/ijms25116136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
The placenta plays a key role in several adverse obstetrical outcomes, such as preeclampsia, intrauterine growth restriction and gestational diabetes mellitus. The early identification of at-risk pregnancies could significantly improve the management, therapy and prognosis of these pregnancies, especially if these at-risk pregnancies are identified in the first trimester. The aim of this review was to summarize the possible biomarkers that can be used to diagnose early placental dysfunction and, consequently, at-risk pregnancies. We divided the biomarkers into proteins and non-proteins. Among the protein biomarkers, some are already used in clinical practice, such as the sFLT1/PLGF ratio or PAPP-A; others are not yet validated, such as HTRA1, Gal-3 and CD93. In the literature, many studies analyzed the role of several protein biomarkers, but their results are contrasting. On the other hand, some non-protein biomarkers, such as miR-125b, miR-518b and miR-628-3p, seem to be linked to an increased risk of complicated pregnancy. Thus, a first trimester heterogeneous biomarkers panel containing protein and non-protein biomarkers may be more appropriate to identify and discriminate several complications that can affect pregnancies.
Collapse
Affiliation(s)
- Martina Cristodoro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milano, Italy; (M.C.)
| | - Martina Messa
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milano, Italy; (M.C.)
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126 Ancona, Italy
| | | | - Annalisa Inversetti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milano, Italy; (M.C.)
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milano, Italy; (M.C.)
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
56
|
Louwen F, Kreis NN, Ritter A, Yuan J. Maternal obesity and placental function: impaired maternal-fetal axis. Arch Gynecol Obstet 2024; 309:2279-2288. [PMID: 38494514 PMCID: PMC11147848 DOI: 10.1007/s00404-024-07462-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
The prevalence of maternal obesity rapidly increases, which represents a major public health concern worldwide. Maternal obesity is characteristic by metabolic dysfunction and chronic inflammation. It is associated with health problems in both mother and offspring. Increasing evidence indicates that the placenta is an axis connecting maternal obesity with poor outcomes in the offspring. In this brief review, we have summarized the current data regarding deregulated placental function in maternal obesity. The data show that maternal obesity induces numerous placental defects, including lipid and glucose metabolism, stress response, inflammation, immune regulation and epigenetics. These placental defects affect each other and result in a stressful intrauterine environment, which transduces and mediates the adverse effects of maternal obesity to the fetus. Further investigations are required to explore the exact molecular alterations in the placenta in maternal obesity, which may pave the way to develop specific interventions for preventing epigenetic and metabolic programming in the fetus.
Collapse
Affiliation(s)
- Frank Louwen
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor Stern-Kai 7, 60590, Frankfurt, Germany
| | - Nina-Naomi Kreis
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor Stern-Kai 7, 60590, Frankfurt, Germany
| | - Andreas Ritter
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor Stern-Kai 7, 60590, Frankfurt, Germany
| | - Juping Yuan
- Obstetrics and Prenatal Medicine, Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Theodor Stern-Kai 7, 60590, Frankfurt, Germany.
| |
Collapse
|
57
|
Wang J, Liu W, Zhuang Y, Yang J, Zhao Y, Hong A, Du J, Kong H, Wang J, Jiang Y, Wang Y. Influenza A virus infection disrupts the function of syncytiotrophoblast cells and contributes to adverse pregnancy outcomes. J Med Virol 2024; 96:e29687. [PMID: 38783821 DOI: 10.1002/jmv.29687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/23/2024] [Accepted: 05/12/2024] [Indexed: 05/25/2024]
Abstract
Pregnancy heightens susceptibility to influenza A virus (IAV) infection, thereby increasing the risk of severe pneumonia and maternal mortality. It also raises the chances of adverse outcomes in offspring, such as fetal growth restriction, preterm birth, miscarriage, and stillbirth in offsprings. However, the underlying mechanisms behind these effects remain largely unknown. Syncytiotrophoblast cells, crucial in forming the placental barrier, nutrient exchange and hormone secretion, have not been extensively studied for their responses to IAV. In our experiment, we used Forskolin-treated BeWo cells to mimic syncytiotrophoblast cells in vitro, and infected them with H1N1, H5N1 and H7N9 virus stains. Our results showed that syncytiotrophoblast cells, with their higher intensity of sialic acid receptors, strongly support IAV infection and replication. Notably, high-dose viral infection and prolonged exposure resulted in a significant decrease in fusion index, as well as gene and protein expression levels associated with trophoblast differentiation, β-human chorionic gonadotropin secretion, estrogen and progesterone biosynthesis, and nutrient transport. In pregnant BALB/c mice infected with the H1N1 virus, we observed significant decreases in trophoblast differentiation and hormone secretion gene expression levels. IAV infection also resulted in preterm labor, fetal growth restriction, and increased maternal and fetal morbidity and mortality. Our findings indicate that IAV infection in syncytiotrophoblastic cells can result in adverse pregnancy outcomes by altering trophoblast differentiation, suppressing of β-hCG secretion, and disrupting placental barrier function.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenyu Liu
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yichao Zhuang
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Jiaxin Yang
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yetian Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Aihui Hong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jingjing Du
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huihui Kong
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Jingfei Wang
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yongping Jiang
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yan Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
58
|
Tamil Barathi P, Mohanapriya A. Pre-eclampsia: Re-visiting pathophysiology, role of immune cells, biomarker identification and recent advances in its management. J Reprod Immunol 2024; 163:104236. [PMID: 38555746 DOI: 10.1016/j.jri.2024.104236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Pre-eclampsia (PE) is a hypertension condition that occurs exclusively during pregnancy and has the potential to impact nearly all organ systems. It is estimated to complicate approximately 2-8% of pregnancies worldwide. PE is a prominent medical disorder that poses a significant risk to pregnant mothers and their infants. This review commences by giving the most up-to- date concepts about the pathophysiology of PE. The condition involves atypical infiltration of trophoblast cells into the spiral arteries of the decidua and myometrium, resulting in an insufficient establishment of proper blood flow between the uterus and placenta. The aberrant activation of natural killer (NK) cells in both the peripheral blood and the decidua has been identified as one of the contributing factors to the development of PE. The strong evidence for the genetic etiology of PE is provided by the association between maternal killer cell immunoglobulin-like receptor (KIR) and Human Leukocyte Antigen (HLA-C) in trophoblast cells. Recent observations provide evidence that changes in the expression of anti-angiogenic factors in the placenta are the underlying cause of the clinical symptoms associated with the condition. This review also provides a comprehensive overview of the latest advancements in understanding the underlying causes of PE. It specifically highlights the emergence of new diagnostic biomarkers and their potential implications for therapeutic interventions in managing this medical condition.
Collapse
Affiliation(s)
- Palanisamy Tamil Barathi
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| | - Arumugam Mohanapriya
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| |
Collapse
|
59
|
Cinzori ME, Pacyga DC, Rosas L, Whalen J, Smith S, Park JS, Geiger SD, Gardiner JC, Braun JM, Schantz SL, Strakovsky RS. Associations of per- and polyfluoroalkyl substances with maternal metabolic and inflammatory biomarkers in early-to-mid-pregnancy. ENVIRONMENTAL RESEARCH 2024; 250:118434. [PMID: 38346483 PMCID: PMC11102845 DOI: 10.1016/j.envres.2024.118434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/02/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFAS) can disrupt metabolism. Early-to-mid pregnancy is characterized by amplified metabolic processes and inflammation to support maternal adaptations and fetal growth. Thus, we cross-sectionally evaluated whether PFAS are individually and jointly associated with these processes in early-to-mid pregnancy. METHODS Pregnant Illinois women (n = 452) provided fasted blood samples at median 17 weeks gestation. We quantified serum perfluorononanoic (PFNA), perfluorooctane sulfonic (PFOS), perfluorooctanoic (PFOA), methyl-perfluorooctane sulfonamide acetic acid (Me-PFOSA-AcOH), perfluorohexanesulfonic (PFHxS), perfluorodecanoic (PFDeA), and perfluoroundecanoic (PFUdA) acid. Key outcomes were plasma glucose, insulin, C-peptide, insulin-like growth factor 1 (IGF-1), adiponectin, leptin, triglycerides, free fatty acids, total cholesterol, high-density lipoprotein (HDL) cholesterol, C-reactive protein, tumor necrosis factor alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and interleukin 6. We calculated homeostatic model assessment for insulin resistance (HOMA-IR), low-density lipoprotein (LDL) cholesterol, and very low-density lipoprotein (VLDL). We evaluated associations of PFAS with each metabolic/inflammatory biomarker individually using covariate-adjusted linear regression and jointly using quantile-based g-computation. RESULTS In linear regression, all PFAS (except Me-PFOSA-AcOH) were negatively associated with insulin, HOMA-IR, and leptin, whereas all PFAS were positively associated with HDL cholesterol. We also observed negative associations of some PFAS with TNF-α and MCP-1; positive associations with adiponectin and total cholesterol also emerged. Additionally, PFOS was positively, whereas Me-PFOSA-AcOH was negatively, associated with triglycerides and VLDL. Each 25% increase in the PFAS mixture was associated with -31.3% lower insulin (95%CI: -45.8, -12.9), -31.9% lower HOMA-IR (95%CI: -46.4, -13.4), and -9.4% lower leptin (95%CI: -17.3, -0.8), but 7.4% higher HDL cholesterol (95%CI: 4.6, 10.3). For most outcomes, the major contributors to the PFAS mixture often differed compared to single-PFAS analyses. IMPLICATIONS Individual and joint PFAS exposures were associated with markers of maternal metabolism and inflammation in pregnancy. Further investigation is needed to elucidate possible mechanisms and consequences of these findings.
Collapse
Affiliation(s)
- Maria E Cinzori
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, 48824, USA
| | - Diana C Pacyga
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA
| | - Libeth Rosas
- The Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, 61801, USA
| | - Jason Whalen
- Michigan Diabetes Research Center Chemistry Laboratory, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Sabrina Smith
- Environmental Chemistry Laboratory, Department of Toxic Substances Control, California Environmental Protection Agency, Berkeley, CA, 94710, USA
| | - June-Soo Park
- Environmental Chemistry Laboratory, Department of Toxic Substances Control, California Environmental Protection Agency, Berkeley, CA, 94710, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, CA 94158, USA
| | - Sarah D Geiger
- The Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, 61801, USA; Department of Kinesiology and Community Health, University of Illinois, Urbana-Champaign, IL 61801, USA
| | - Joseph C Gardiner
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, 48824, USA
| | - Joseph M Braun
- Department of Epidemiology, Brown University, Providence, RI, 02912, USA
| | - Susan L Schantz
- The Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, IL, 61801, USA; Department of Comparative Biosciences, University of Illinois, Urbana-Champaign, IL 61802, USA
| | - Rita S Strakovsky
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, 48824, USA; Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
60
|
Cromb D, Slator PJ, Hall M, Price A, Alexander DC, Counsell SJ, Hutter J. Advanced magnetic resonance imaging detects altered placental development in pregnancies affected by congenital heart disease. Sci Rep 2024; 14:12357. [PMID: 38811636 PMCID: PMC11136986 DOI: 10.1038/s41598-024-63087-8] [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: 01/17/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024] Open
Abstract
Congenital heart disease (CHD) is the most common congenital malformation and is associated with adverse neurodevelopmental outcomes. The placenta is crucial for healthy fetal development and placental development is altered in pregnancy when the fetus has CHD. This study utilized advanced combined diffusion-relaxation MRI and a data-driven analysis technique to test the hypothesis that placental microstructure and perfusion are altered in CHD-affected pregnancies. 48 participants (36 controls, 12 CHD) underwent 67 MRI scans (50 control, 17 CHD). Significant differences in the weighting of two independent placental and uterine-wall tissue components were identified between the CHD and control groups (both pFDR < 0.001), with changes most evident after 30 weeks gestation. A significant trend over gestation in weighting for a third independent tissue component was also observed in the CHD cohort (R = 0.50, pFDR = 0.04), but not in controls. These findings add to existing evidence that placental development is altered in CHD. The results may reflect alterations in placental perfusion or the changes in fetal-placental flow, villous structure and maturation that occur in CHD. Further research is needed to validate and better understand these findings and to understand the relationship between placental development, CHD, and its neurodevelopmental implications.
Collapse
Affiliation(s)
- Daniel Cromb
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK
- Centre for Medical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Paddy J Slator
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
- School of Computer Science and Informatics, Cardiff University, Cardiff, UK
- Cardiff University Brain Research Imaging Centre, School of Psychology, Cardiff University, Cardiff, UK
| | - Megan Hall
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK
| | - Anthony Price
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK
- Centre for Medical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Daniel C Alexander
- Centre for Medical Image Computing, Department of Computer Science, University College London, London, UK
| | - Serena J Counsell
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK.
- Centre for Medical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.
| | - Jana Hutter
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, London, SE1 7EH, UK
- Centre for Medical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
- Smart Imaging Lab, Radiological Institute, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
61
|
Li H, Miao D, Hu H, Xue P, Zhou K, Mao Z. Titanium Dioxide Nanoparticles Induce Maternal Preeclampsia-like Syndrome and Adverse Birth Outcomes via Disrupting Placental Function in SD Rats. TOXICS 2024; 12:367. [PMID: 38787146 PMCID: PMC11125676 DOI: 10.3390/toxics12050367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
The escalating utilization of titanium dioxide nanoparticles (TiO2 NPs) in everyday products has sparked concerns regarding their potential hazards to pregnant females and their offspring. To address these concerns and shed light on their undetermined adverse effects and mechanisms, we established a pregnant rat model to investigate the impacts of TiO2 NPs on both maternal and offspring health and to explore the underlying mechanisms of those impacts. Pregnant rats were orally administered TiO2 NPs at a dose of 5 mg/kg body weight per day from GD5 to GD18 during pregnancy. Maternal body weight, organ weight, and birth outcomes were monitored and recorded. Maternal pathological changes were examined by HE staining and TEM observation. Maternal blood pressure was assessed using a non-invasive blood analyzer, and the urinary protein level was determined using spot urine samples. Our findings revealed that TiO2 NPs triggered various pathological alterations in maternal liver, kidney, and spleen, and induced maternal preeclampsia-like syndrome, as well as leading to growth restriction in the offspring. Further examination unveiled that TiO2 NPs hindered trophoblastic cell invasion into the endometrium via the promotion of autophagy. Consistent hypertension and proteinuria resulted from the destroyed the kidney GBM. In total, an exposure to TiO2 NPs during pregnancy might increase the risk of human preeclampsia through increased maternal arterial pressure and urinary albumin levels, as well as causing fetal growth restriction in the offspring.
Collapse
Affiliation(s)
- Haixin Li
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China; (H.L.); (H.H.); (P.X.)
| | - Dandan Miao
- Huai’an Center for Disease Control and Prevention, Huai’an 223001, China;
| | - Haiting Hu
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China; (H.L.); (H.H.); (P.X.)
| | - Pingping Xue
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China; (H.L.); (H.H.); (P.X.)
| | - Kun Zhou
- State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing 211100, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211100, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhilei Mao
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China; (H.L.); (H.H.); (P.X.)
- State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing 211100, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211100, China
| |
Collapse
|
62
|
Osman HC, Moreno R, Rose D, Rowland ME, Ciernia AV, Ashwood P. Impact of maternal immune activation and sex on placental and fetal brain cytokine and gene expression profiles in a preclinical model of neurodevelopmental disorders. J Neuroinflammation 2024; 21:118. [PMID: 38715090 PMCID: PMC11077729 DOI: 10.1186/s12974-024-03106-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/21/2024] [Indexed: 05/12/2024] Open
Abstract
Maternal inflammation during gestation is associated with a later diagnosis of neurodevelopmental disorders including autism spectrum disorder (ASD). However, the specific impact of maternal immune activation (MIA) on placental and fetal brain development remains insufficiently understood. This study aimed to investigate the effects of MIA by analyzing placental and brain tissues obtained from the offspring of pregnant C57BL/6 dams exposed to polyinosinic: polycytidylic acid (poly I: C) on embryonic day 12.5. Cytokine and mRNA content in the placenta and brain tissues were assessed using multiplex cytokine assays and bulk-RNA sequencing on embryonic day 17.5. In the placenta, male MIA offspring exhibited higher levels of GM-CSF, IL-6, TNFα, and LT-α, but there were no differences in female MIA offspring. Furthermore, differentially expressed genes (DEG) in the placental tissues of MIA offspring were found to be enriched in processes related to synaptic vesicles and neuronal development. Placental mRNA from male and female MIA offspring were both enriched in synaptic and neuronal development terms, whereas females were also enriched for terms related to excitatory and inhibitory signaling. In the fetal brain of MIA offspring, increased levels of IL-28B and IL-25 were observed with male MIA offspring and increased levels of LT-α were observed in the female offspring. Notably, we identified few stable MIA fetal brain DEG, with no male specific difference whereas females had DEG related to immune cytokine signaling. Overall, these findings support the hypothesis that MIA contributes to the sex- specific abnormalities observed in ASD, possibly through altered neuron developed from exposure to inflammatory cytokines. Future research should aim to investigate how interactions between the placenta and fetal brain contribute to altered neuronal development in the context of MIA.
Collapse
Affiliation(s)
- Hadley C Osman
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Rachel Moreno
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Destanie Rose
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA
| | - Megan E Rowland
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Annie Vogel Ciernia
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, Davis, CA, USA.
- The M.I.N.D. Institute, University of California at Sacramento, Sacramento, CA, USA.
| |
Collapse
|
63
|
Bakoyan Z, Cao Y, Hansson SR, Karlsson JP, Lodefalk M. Childhood atopic disorders in relation to placental changes-A systematic review and meta-analysis. Pediatr Allergy Immunol 2024; 35:e14141. [PMID: 38773752 DOI: 10.1111/pai.14141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/24/2024]
Abstract
Fetal programming may arise from prenatal exposure and increase the risk of diseases later in life, potentially mediated by the placenta. The objective of this systematic review was to summarize and critically evaluate publications describing associations between human placental changes and risk of atopic disorders during childhood. The review adhered to the Preferred Reporting Items for Systematic Reviews and Meta-analysis guidelines. The inclusion criteria were original research articles or case reports written in English describing a human placental change in relation to disease occurring in offspring during childhood. The MEDLINE and EMBASE databases were searched for eligible studies. Risk of bias (RoB) was assessed using the ROBINS-I tool. The results were pooled both in a narrative way and by a meta-analysis. Nineteen studies were included (n = 12,997 participants). All studies had an overall serious RoB, and publication bias could not be completely ruled out. However, five studies showed that histological chorioamnionitis in preterm-born children was associated with asthma-related problems (pooled odds ratio = 3.25 (95% confidence interval = 2.22-4.75)). In term-born children, a large placenta (≥750 g) increased the risk of being prescribed anti-asthma medications during the first year of life. Placental histone acetylation, DNA methylation, and gene expression differences were found to be associated with different atopic disorders in term-born children. There is some evidence supporting the idea that the placenta can mediate an increased risk of atopic disorders in children. However, further studies are needed to validate the findings, properly control for confounders, and examine potential mechanisms.
Collapse
Affiliation(s)
- Zaki Bakoyan
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Yang Cao
- Clinical Epidemiology and Biostatistics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Stefan R Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Science Lund, Lund University, Lund, Sweden
| | | | - Maria Lodefalk
- University Health Care Research Center, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Pediatrics, School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
64
|
Santacruz-Márquez R, Neff AM, Mourikes VE, Fletcher EJ, Flaws JA. The effects of inhaled pollutants on reproduction in marginalized communities: a contemporary review. Inhal Toxicol 2024; 36:286-303. [PMID: 37075037 PMCID: PMC10584991 DOI: 10.1080/08958378.2023.2197941] [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: 10/14/2022] [Accepted: 03/25/2023] [Indexed: 04/20/2023]
Abstract
Important differences in health that are closely linked with social disadvantage exist within and between countries. According to the World Health Organization, life expectancy and good health continue to increase in many parts of the world, but fail to improve in other parts of the world, indicating that differences in life expectancy and health arise due to the circumstances in which people grow, live, work, and age, and the systems put in place to deal with illness. Marginalized communities experience higher rates of certain diseases and more deaths compared to the general population, indicating a profound disparity in health status. Although several factors place marginalized communities at high risk for poor health outcomes, one important factor is exposure to air pollutants. Marginalized communities and minorities are exposed to higher levels of air pollutants than the majority population. Interestingly, a link exists between air pollutant exposure and adverse reproductive outcomes, suggesting that marginalized communities may have increased reproductive disorders due to increased exposure to air pollutants compared to the general population. This review summarizes different studies showing that marginalized communities have higher exposure to air pollutants, the types of air pollutants present in our environment, and the associations between air pollution and adverse reproductive outcomes, focusing on marginalized communities.
Collapse
Affiliation(s)
| | - Alison M. Neff
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign
| | | | - Endia J. Fletcher
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign
| | - Jodi A. Flaws
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign
| |
Collapse
|
65
|
Zhao D, Cheng T, Hu D, Xu X, Zhang F, Yu R, Li H, Wen P, Chen L, Fu M, Yang H, Zhang H, Yao J, Jin L. Maternal periodontal diseases affect the leukocyte profiles of umbilical cord blood: A cohort study. Oral Dis 2024; 30:2533-2545. [PMID: 37485723 DOI: 10.1111/odi.14683] [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: 04/27/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/25/2023]
Abstract
AIM This study evaluated the connection of periodontal status with the leukocyte profiles of maternal peripheral blood (MPB) and umbilical cord blood (UCB). MATERIALS AND METHODS Ninety-nine pregnant females were recruited, and their data were collected via questionnaire and from medical records, including demographics, systemic conditions, complete blood count (CBC) and C-reaction protein (CRP) level in MPB. Full-mouth periodontal assessment was performed. CBC and CRP levels in UCB were measured after parturition. RESULTS All subjects and their neonates were generally healthy. 30.3% of the participants presented with periodontal health condition, whereas 69.7% had different severities of periodontal diseases. The counts/percentages of eosinophils and monocytes in UCB from the subjects with periodontal diseases elevated, and the percentage of neutrophils decreased referencing to that from the counterparts (p < 0.05). There were positive correlations for total leukocyte count, neutrophils and lymphocytes counts/percentages in MPB and UCB among the periodontally healthy subjects (r > 0.4, p < 0.05), but such findings did not exist in those with periodontal diseases. Moreover, periodontal diseases independently accounted for the counts/percentages of neutrophils and eosinophils in UCB after controlling confounders in four testing models (ANCOVA, p < 0.05). CONCLUSION Maternal periodontal diseases could to some extent disturb the leukocyte profiles of umbilical cord blood.
Collapse
Affiliation(s)
- Dan Zhao
- Department of Implant Dentistry, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
- Division of Periodontology & Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Tianfan Cheng
- Division of Periodontology & Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Dangli Hu
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Xiaoyi Xu
- Division of Science & Education, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Feng Zhang
- Division of Stomatology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Rong Yu
- Division of Science & Education, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Huijun Li
- Division of Stomatology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Ping Wen
- Division of Science & Education, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Lihua Chen
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Mali Fu
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Hong Yang
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Hanyu Zhang
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Jilong Yao
- Division of Obstetrics & Gynecology, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Lijian Jin
- Division of Periodontology & Implant Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
66
|
Ma J, Liu M, Chen Z, Liu S, Yang H, Duan M. NANOG regulate the JAK/STAT3 pathway to promote trophoblast cell migration and epithelial-mesenchymal transition (EMT) in hypertensive disorders of pregnancy (HDP) through protein interaction with CDK1. Am J Reprod Immunol 2024; 91:e13863. [PMID: 38796740 DOI: 10.1111/aji.13863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 04/07/2024] [Accepted: 04/26/2024] [Indexed: 05/28/2024] Open
Abstract
PROBLEM Hypertensive disorders of pregnancy (HDP) are a common pregnancy disease. NANOG and Cyclin-dependent kinase 1 (CDK1) are essential for regulating the function of cell proliferation and apoptosis. However, the mechanism of action in HDP is yet unclear. METHOD The microarray dataset GSE6573 was downloaded from the GEO database. Emt-related gene set was downloaded from Epithelial-Mesenchymal Transition gene database 2.0 were screened differentially expressed genes by bioinformatics analysis. Pathway Commons and Scansite 4.0 databases were used to predict the interaction between proteins. Placental tissue samples were collected from HDP patients and patients with uneventful pregnancies. RT-qPCR, Western blot and immunohistochemistry were used to detect the expression of NANOG, CDK1, MMP-2, MMP-9, EMT markers and the JAK/STAT3 pathway proteins. Transfection NANOG overexpression/knockdown, and CDK1 knockdown into the human chorionic trophoblast cells (HTR-8/Svneo). CCK-8, Transwell and Wound-healing assay were used to evaluate cell proliferation, invasion and migration. CO-IP and GST pull-down assays were used to confirm the protein interaction. RESULTS A total obtained seven EMT-related differentially expressed genes, wherein NANOG, NODAL and LIN28A had protein interaction. In the HDP patients' tissue found that NANOG and CDK1 had lower expression. NANOG overexpression promoted HTR-8/Svneo proliferation, migration and EMT, while NANOG knockdown had the opposite effect. Further a protein interaction between STAT3 and CDK1 with NANOG. NANOG overexpression downregulated the JAK/STAT3 pathway to promote HTR-8/Svneo proliferation, migration and EMT, which was reversed by CDK1 knockdown. CONCLUSIONS NANOG downregulated the JAK/STAT3 pathway to promote trophoblast cell proliferation, migration and EMT through protein interaction with CDK1.
Collapse
Affiliation(s)
- Jing Ma
- Department of Reproduction and Genetics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Mingchang Liu
- Yunnan Maternal and Child Health Care Hospital, Kunming, Yunnan, China
- Kunming Medical University, Kunming, Yunnan, China
| | - Zhuo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Shiyang Liu
- Kunming Medical University, Kunming, Yunnan, China
| | - Huijuan Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Mengjia Duan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
67
|
Wang Y, Chen Z, Li J, Wan T, Hu R, Zhang L, Qin L, Zang L, Gu W, Chen R, Liu C, Li R. Gestational exposure to PM 2.5 disrupts fetal development by suppressing placental trophoblast syncytialization via progranulin/mTOR signaling. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 921:171101. [PMID: 38387595 DOI: 10.1016/j.scitotenv.2024.171101] [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: 01/08/2024] [Revised: 02/06/2024] [Accepted: 02/18/2024] [Indexed: 02/24/2024]
Abstract
Recent epidemiological and animal studies have indicated that ambient fine particulate matter (PM2.5) exposure during pregnancy is closely associated with intrauterine growth restriction (IUGR). However, the underlying mechanisms remain to be revealed. In this study, we found that gestational exposure to PM2.5 significantly decreased fetal weight and crown-rump length in mice, accompanied by insufficient placental trophoblast syncytialization and increased expression of progranulin (PGRN) in mice placenta. Administering PGRN neutralizing antibody to pregnant mice alleviated growth restriction and insufficient placental trophoblast syncytialization caused by PM2.5, accompanied with suppressed activation of the mTOR signaling pathway. Furthermore, in vitro experiments using human placental BeWo cells showed that 10 μg·mL-1 PM2.5 activated PGRN/mTOR signaling and suppressed forskolin-induced cell fusion, which was blocked by knockdown of PGRN. Taken together, our results demonstrated that PM2.5 exposure during pregnancy inhibited placental trophoblast syncytialization by activating PGRN/mTOR signaling, leading to abnormal placental development and IUGR. This study reveals a novel mechanism underlying the developmental toxicity of PM2.5 exposure during pregnancy.
Collapse
Affiliation(s)
- Yirun Wang
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuan Chen
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Li
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Teng Wan
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Renjie Hu
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zhang
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Qin
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Zang
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weijia Gu
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rucheng Chen
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Ran Li
- School of Public Health, Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
68
|
Ho SJ, Chaput D, Sinkey RG, Garces AH, New EP, Okuka M, Sang P, Arlier S, Semerci N, Steffensen TS, Rutherford TJ, Alsina AE, Cai J, Anderson ML, Magness RR, Uversky VN, Cummings DAT, Tsibris JCM. Proteomic studies of VEGFR2 in human placentas reveal protein associations with preeclampsia, diabetes, gravidity, and labor. Cell Commun Signal 2024; 22:221. [PMID: 38594674 PMCID: PMC11003095 DOI: 10.1186/s12964-024-01567-0] [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: 10/21/2023] [Accepted: 03/09/2024] [Indexed: 04/11/2024] Open
Abstract
VEGFR2 (Vascular endothelial growth factor receptor 2) is a central regulator of placental angiogenesis. The study of the VEGFR2 proteome of chorionic villi at term revealed its partners MDMX (Double minute 4 protein) and PICALM (Phosphatidylinositol-binding clathrin assembly protein). Subsequently, the oxytocin receptor (OT-R) and vasopressin V1aR receptor were detected in MDMX and PICALM immunoprecipitations. Immunogold electron microscopy showed VEGFR2 on endothelial cell (EC) nuclei, mitochondria, and Hofbauer cells (HC), tissue-resident macrophages of the placenta. MDMX, PICALM, and V1aR were located on EC plasma membranes, nuclei, and HC nuclei. Unexpectedly, PICALM and OT-R were detected on EC projections into the fetal lumen and OT-R on 20-150 nm clusters therein, prompting the hypothesis that placental exosomes transport OT-R to the fetus and across the blood-brain barrier. Insights on gestational complications were gained by univariable and multivariable regression analyses associating preeclampsia with lower MDMX protein levels in membrane extracts of chorionic villi, and lower MDMX, PICALM, OT-R, and V1aR with spontaneous vaginal deliveries compared to cesarean deliveries before the onset of labor. We found select associations between higher MDMX, PICALM, OT-R protein levels and either gravidity, diabetes, BMI, maternal age, or neonatal weight, and correlations only between PICALM-OT-R (p < 2.7 × 10-8), PICALM-V1aR (p < 0.006), and OT-R-V1aR (p < 0.001). These results offer for exploration new partnerships in metabolic networks, tissue-resident immunity, and labor, notably for HC that predominantly express MDMX.
Collapse
Grants
- Department of Obstetrics and Gynecology, University of South Florida
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida
- Lisa Muma Weitz Microscopy Laboratory, University of South Florida
- Department of Chemistry, University of South Florida
- Tampa General Hospital, Tampa, Florida
- Teasley Foundation
- Department of Molecular Medicine, University of South Florida
- Department of Biology, University of Florida
- Emerging Pathogens Institute, University of Florida
Collapse
Affiliation(s)
- Shannon J Ho
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Dale Chaput
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Rachel G Sinkey
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Amanda H Garces
- Lisa Muma Weitz Microscopy Laboratory, University of South Florida, Tampa, FL, USA
| | - Erika P New
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Maja Okuka
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Peng Sang
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Sefa Arlier
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Nihan Semerci
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | | | - Thomas J Rutherford
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
- Cancer Center, Tampa General Hospital, Tampa, FL, USA
| | - Angel E Alsina
- Transplant Surgery Center, Tampa General Hospital, Tampa, FL, USA
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL, USA
| | - Matthew L Anderson
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
- Cancer Center, Tampa General Hospital, Tampa, FL, USA
| | - Ronald R Magness
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA
| | - Vladimir N Uversky
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA
| | - Derek A T Cummings
- Department of Biology and Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA
| | - John C M Tsibris
- Department of Obstetrics and Gynecology, University of South Florida, Tampa, FL, USA.
- Department of Molecular Medicine, University of South Florida, Tampa, FL, USA.
| |
Collapse
|
69
|
Calvo MJ, Parra H, Santeliz R, Bautista J, Luzardo E, Villasmil N, Martínez MS, Chacín M, Cano C, Checa-Ros A, D'Marco L, Bermúdez V, De Sanctis JB. The Placental Role in Gestational Diabetes Mellitus: A Molecular Perspective. TOUCHREVIEWS IN ENDOCRINOLOGY 2024; 20:10-18. [PMID: 38812661 PMCID: PMC11132656 DOI: 10.17925/ee.2024.20.1.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/01/2023] [Indexed: 05/31/2024]
Abstract
During pregnancy, women undergo several metabolic changes to guarantee an adequate supply of glucose to the foetus. These metabolic modifications develop what is known as physiological insulin resistance. When this process is altered, however, gestational diabetes mellitus (GDM) occurs. GDM is a multifactorial disease, and genetic and environmental factors play a crucial role in its aetiopathogenesis. GDM has been linked to both macroscopic and molecular alterations in placental tissues that affect placental physiology. This review summarizes the role of the placenta in the development of GDM from a molecular perspective, including hormonal and pro-inflammatory changes. Inflammation and hormonal imbalance, the characteristics dominating the GDM microenvironment, are responsible for placental changes in size and vascularity, leading to dysregulation in maternal and foetal circulations and to complications in the newborn. In conclusion, since the hormonal mechanisms operating in GDM have not been fully elucidated, more research should be done to improve the quality of life of patients with GDM and their future children.
Collapse
Affiliation(s)
- María José Calvo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Heliana Parra
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Raquel Santeliz
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Jordan Bautista
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Eliana Luzardo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Nelson Villasmil
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - María Sofía Martínez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Maricamen Chacín
- Facultad de Ciencias de la Salud, Barranquilla, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Ana Checa-Ros
- Research Group on Cardiorenal and Metabolic Diseases, Departamento de Medicina y Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
- School of Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Luis D'Marco
- Research Group on Cardiorenal and Metabolic Diseases, Departamento de Medicina y Cirugía, Facultad de Ciencias de la Salud, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia, Spain
| | - Valmore Bermúdez
- Facultad de Ciencias de la Salud, Barranquilla, Universidad Simón Bolívar, Barranquilla, Colombia
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
70
|
Tuomivaara ST, Goin DE, Fisher SJ, Hall SC, Mattis AN, Den Besten PK. Fluoride-related changes in the fetal cord blood proteome; a pilot study. RESEARCH SQUARE 2024:rs.3.rs-3995767. [PMID: 38464284 PMCID: PMC10925477 DOI: 10.21203/rs.3.rs-3995767/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Fluoride exposure during pregnancy has been associated with various effects on offspring, including changes in behavior and IQ. To provide clues to possible mechanisms by which fluoride affects human fetal development, we completed proteomic analyses of cord blood serum collected from second-trimester pregnant women residing in Northern California with either high or low fluoride exposure, as identified by maternal serum fluoride concentrations. Objective To identify changes in cord blood proteins associated with maternal serum fluoride concentration in pregnant women living in Northern California. Methods The proteomes of 19 archived second-trimester cord blood samples representing highest and lowest serum fluoride concentrations from a cohort of 48 women living in Northern California, previously analyzed for serum, urine and amniotic fluoride concentrations, were characterized by mass spectrometry. Proteins highly correlated to maternal serum fluoride concentrations were identified, and further compared in a group of samples from women with the highest serum fluoride to the group with the lowest maternal serum fluoride concentrations. Results Nine cord blood proteins were significantly correlated with maternal serum fluoride concentrations. Six of these proteins, including apolipoprotein B-100, delta homolog 1, coagulation factor X, mimecan, plasma kallikrein, and vasorin, were significantly decreased in the cord blood from women with the highest serum fluoride levels. Conclusion Changes in the relative amounts of second trimester cord blood proteins included proteins associated with the development of the fetal hematopoetic system.
Collapse
Affiliation(s)
| | - Dana E Goin
- University of California, San Francisco San Francisco
| | | | - Steven C Hall
- University of California, San Francisco San Francisco
| | - Aras N Mattis
- University of California, San Francisco San Francisco
| | | |
Collapse
|
71
|
Fernandes LM, Lorigo M, Cairrao E. Relationship between Androgens and Vascular and Placental Function during Pre-eclampsia. Curr Issues Mol Biol 2024; 46:1668-1693. [PMID: 38534724 DOI: 10.3390/cimb46030108] [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: 01/24/2024] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 03/28/2024] Open
Abstract
Hypertensive disorders of pregnancy (HDP) represent a substantial risk to maternal and fetal health. Emerging evidence suggests an association between testosterone and pre-eclampsia (PE), potentially mediated through androgen receptors (AR). Nevertheless, the mechanism driving this association is yet to be elucidated. On the other hand, reports of transgender men's pregnancies offer a limited and insightful opportunity to understand the role of high androgen levels in the development of HDP. In this sense, a literature review was performed from a little over 2 decades (1998-2022) to address the association of testosterone levels with the development of HDP. Furthermore, this review addresses the case of transgender men for the first time. The main in vitro outcomes reveal placenta samples with greater AR mRNA expression. Moreover, ex vivo studies show that testosterone-induced vasorelaxation impairment promotes hypertension. Epidemiological data point to greater testosterone levels in blood samples during PE. Studies with transgender men allow us to infer that exogenous testosterone administration can be considered a risk factor for PE and that the administration of testosterone does not affect fetal development. Overall, all studies analyzed suggested that high testosterone levels are associated with PE.
Collapse
Affiliation(s)
- Lara M Fernandes
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal
- FCS-UBI, Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Margarida Lorigo
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal
- FCS-UBI, Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Elisa Cairrao
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal
- FCS-UBI, Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| |
Collapse
|
72
|
Casazza W, Inkster AM, Del Gobbo GF, Yuan V, Delahaye F, Marsit C, Park YP, Robinson WP, Mostafavi S, Dennis JK. Sex-dependent placental methylation quantitative trait loci provide insight into the prenatal origins of childhood onset traits and conditions. iScience 2024; 27:109047. [PMID: 38357671 PMCID: PMC10865402 DOI: 10.1016/j.isci.2024.109047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/19/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Molecular quantitative trait loci (QTLs) allow us to understand the biology captured in genome-wide association studies (GWASs). The placenta regulates fetal development and shows sex differences in DNA methylation. We therefore hypothesized that placental methylation QTL (mQTL) explain variation in genetic risk for childhood onset traits, and that effects differ by sex. We analyzed 411 term placentas from two studies and found 49,252 methylation (CpG) sites with mQTL and 2,489 CpG sites with sex-dependent mQTL. All mQTL were enriched in regions that typically affect gene expression in prenatal tissues. All mQTL were also enriched in GWAS results for growth- and immune-related traits, but male- and female-specific mQTL were more enriched than cross-sex mQTL. mQTL colocalized with trait loci at 777 CpG sites, with 216 (28%) specific to males or females. Overall, mQTL specific to male and female placenta capture otherwise overlooked variation in childhood traits.
Collapse
Affiliation(s)
- William Casazza
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital, Vancouver, BC, Canada
- Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Amy M. Inkster
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Giulia F. Del Gobbo
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Victor Yuan
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | | | - Carmen Marsit
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yongjin P. Park
- Department of Statistics, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wendy P. Robinson
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Sara Mostafavi
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital, Vancouver, BC, Canada
- Paul Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Jessica K. Dennis
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital, Vancouver, BC, Canada
- Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
73
|
Luo N, Zhou Y, Chen X, Zhao Y, Hu Y. Screening the optimal housekeeping genes (HKGs) of placenta tissues by RNA-sequence and qRT-PCR throughout gestation in goat (Capra Hircus). Gene 2024; 895:147966. [PMID: 37972698 DOI: 10.1016/j.gene.2023.147966] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/12/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Selection of stable housekeeping genes (HKGs) is very important for accurate calculation of relative expression levels of target genes by quantitative real-time polymerase chain reaction (qRT-PCR). At present, the appropriate HKGs have not been identified in placental tissues throughout the pregnancy of the goat. In our study, 20 HKGs were tentatively selected from RNA-seq data and previous reports. The cycle threshold (Ct) of HKGs was determined by qRT-PCR in trophoblast membrane and cotyledon villus collected from 38 Dazu Black goats on gestation days of 20, 25, 30, 45, 60, 90, 120, and 150 (birth). The expression stability of the HKGs was analyzed by geNorm, Normfinder, Bestkeeper and Delta Ct algorithms, and comprehensively evaluated by ReFinder and ComprFinder. In addition, the optimal HKGs were further verified by placenta-specific genes (SPP1, VEGFA and PAG6). The 16 candidate HKGs (except POP4, TBP, RNF10, UBC) showed a qualified Ct value, less than 28. Among them, YWHAZ, EIF3K and PPIB showed the most stable expression in placental tissues during early, mid-late pregnancy and postpartum, but the least stable expression was B2M at early and mid-late stage, and PPIB at postpartum. After comprehensive analysis, RPLP0, EIF3K and YWHAZ were found to be the most stable placental HKGs throughout pregnancy. The classical HKGs, ACTB, GAPDH and 18S RNA have unstable expressions and even ranked at the bottom of the list from comprehensive index, suggesting an inappropriate for target gene normalization. Taken together, our study confirmed that YWHAZ, EIF3K, HMBS and RPLP0 may be the optimal HKGs in goat placenta at different stage of pregnancy, which provided a valuable reference of HKGs on functional gene expression detection for further research on placenta development and growth in ruminants.
Collapse
Affiliation(s)
- Nanjian Luo
- School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, China; College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yumei Zhou
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Xiaochuan Chen
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China
| | - Yongju Zhao
- College of Animal Science and Technology, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Herbivore Science, Chongqing, 400715, China.
| | - Yu Hu
- School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, China; College of Animal Science and Technology, Southwest University, Chongqing 400715, China; Department of Reproductive Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
74
|
Stroganov S, Harris T, Fellus-Alyagor L, Ben Moyal L, Plitman Mayo R, Golani O, Hirsch D, Ben-Dor S, Brandis A, Mehlman T, Kovo M, Biron-Shental T, Dekel N, Neeman M. The differential regulation of placenta trophoblast bisphosphoglycerate mutase in fetal growth restriction: preclinical study in mice and observational histological study of human placenta. eLife 2024; 13:e82631. [PMID: 38314803 PMCID: PMC10883672 DOI: 10.7554/elife.82631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/25/2024] [Indexed: 02/07/2024] Open
Abstract
Background Fetal growth restriction (FGR) is a pregnancy complication in which a newborn fails to achieve its growth potential, increasing the risk of perinatal morbidity and mortality. Chronic maternal gestational hypoxia, as well as placental insufficiency are associated with increased FGR incidence; however, the molecular mechanisms underlying FGR remain unknown. Methods Pregnant mice were subjected to acute or chronic hypoxia (12.5% O2) resulting in reduced fetal weight. Placenta oxygen transport was assessed by blood oxygenation level dependent (BOLD) contrast magnetic resonance imaging (MRI). The placentae were analyzed via immunohistochemistry and in situ hybridization. Human placentae were selected from FGR and matched controls and analyzed by immunohistochemistry (IHC). Maternal and cord sera were analyzed by mass spectrometry. Results We show that murine acute and chronic gestational hypoxia recapitulates FGR phenotype and affects placental structure and morphology. Gestational hypoxia decreased labyrinth area, increased the incidence of red blood cells (RBCs) in the labyrinth while expanding the placental spiral arteries (SpA) diameter. Hypoxic placentae exhibited higher hemoglobin-oxygen affinity compared to the control. Placental abundance of Bisphosphoglycerate mutase (BPGM) was upregulated in the syncytiotrophoblast and spiral artery trophoblast cells (SpA TGCs) in the murine gestational hypoxia groups compared to the control. Hif1α levels were higher in the acute hypoxia group compared to the control. In contrast, human FGR placentae exhibited reduced BPGM levels in the syncytiotrophoblast layer compared to placentae from healthy uncomplicated pregnancies. Levels of 2,3 BPG, the product of BPGM, were lower in cord serum of human FGR placentae compared to control. Polar expression of BPGM was found in both human and mouse placentae syncytiotrophoblast, with higher expression facing the maternal circulation. Moreover, in the murine SpA TGCs expression of BPGM was concentrated exclusively in the apical cell side, in direct proximity to the maternal circulation. Conclusions This study suggests a possible involvement of placental BPGM in maternal-fetal oxygen transfer, and in the pathophysiology of FGR. Funding This work was supported by the Weizmann Krenter Foundation and the Weizmann - Ichilov (Tel Aviv Sourasky Medical Center) Collaborative Grant in Biomedical Research, by the Minerva Foundation, by the ISF KillCorona grant 3777/19.
Collapse
Affiliation(s)
- Sima Stroganov
- Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Talia Harris
- Chemical Research Support Weizmann Institute of Science, Rehovot, Israel
| | | | - Lital Ben Moyal
- Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Romina Plitman Mayo
- Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Dana Hirsch
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Shifra Ben-Dor
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Alexander Brandis
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Tevie Mehlman
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Kovo
- OBGYN, Meir Medical Center, Kfar Saba, Israel
- Tel Aviv University, School of Medicine, Tel Aviv, Israel
| | - Tal Biron-Shental
- OBGYN, Meir Medical Center, Kfar Saba, Israel
- Tel Aviv University, School of Medicine, Tel Aviv, Israel
| | - Nava Dekel
- Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Neeman
- Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
75
|
O'Brien KA, Gu W, Houck JA, Holzner LMW, Yung HW, Armstrong JL, Sowton AP, Baxter R, Darwin PM, Toledo-Jaldin L, Lazo-Vega L, Moreno-Aramayo AE, Miranda-Garrido V, Shortt JA, Matarazzo CJ, Yasini H, Burton GJ, Moore LG, Simonson TS, Murray AJ, Julian CG. Genomic Selection Signals in Andean Highlanders Reveal Adaptive Placental Metabolic Phenotypes That Are Disrupted in Preeclampsia. Hypertension 2024; 81:319-329. [PMID: 38018457 PMCID: PMC10841680 DOI: 10.1161/hypertensionaha.123.21748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/24/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND The chronic hypoxia of high-altitude residence poses challenges for tissue oxygen supply and metabolism. Exposure to high altitude during pregnancy increases the incidence of hypertensive disorders of pregnancy and fetal growth restriction and alters placental metabolism. High-altitude ancestry protects against altitude-associated fetal growth restriction, indicating hypoxia tolerance that is genetic in nature. Yet, not all babies are protected and placental pathologies associated with fetal growth restriction occur in some Andean highlanders. METHODS We examined placental metabolic function in 79 Andeans (18-45 years; 39 preeclamptic and 40 normotensive) living in La Paz, Bolivia (3600-4100 m) delivered by unlabored Cesarean section. Using a selection-nominated approach, we examined links between putatively adaptive genetic variation and phenotypes related to oxygen delivery or placental metabolism. RESULTS Mitochondrial oxidative capacity was associated with fetal oxygen delivery in normotensive but not preeclamptic placenta and was also suppressed in term preeclamptic pregnancy. Maternal haplotypes in or within 200 kb of selection-nominated genes were associated with lower placental mitochondrial respiratory capacity (PTPRD [protein tyrosine phosphatase receptor-δ]), lower maternal plasma erythropoietin (CPT2 [carnitine palmitoyl transferase 2], proopiomelanocortin, and DNMT3 [DNA methyltransferase 3]), and lower VEGF (vascular endothelial growth factor) in umbilical venous plasma (TBX5 [T-box transcription factor 5]). A fetal haplotype within 200 kb of CPT2 was associated with increased placental mitochondrial complex II capacity, placental nitrotyrosine, and GLUT4 (glucose transporter type 4) protein expression. CONCLUSIONS Our findings reveal novel associations between putatively adaptive gene regions and phenotypes linked to oxygen delivery and placental metabolic function in highland Andeans, suggesting that such effects may be of genetic origin. Our findings also demonstrate maladaptive metabolic mechanisms in the context of preeclampsia, including dysregulation of placental oxygen consumption.
Collapse
Affiliation(s)
- Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Wanjun Gu
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
- Herbert Wertheim School of Public Health and Longevity Sciences (W.G.), University of California San Diego, La Jolla, CA
| | - Julie A Houck
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences (J.A.H., L.G.M.), University of Colorado School of Medicine, Aurora, CO
| | - Lorenz M W Holzner
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Hong Wa Yung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Jenna L Armstrong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Ruby Baxter
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Paula M Darwin
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Lilian Toledo-Jaldin
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Litzi Lazo-Vega
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Any Elena Moreno-Aramayo
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Valquiria Miranda-Garrido
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Jonathan A Shortt
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Christopher J Matarazzo
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Hussna Yasini
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Lorna G Moore
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences (J.A.H., L.G.M.), University of Colorado School of Medicine, Aurora, CO
| | - Tatum S Simonson
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Colleen G Julian
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
76
|
Joseph TT, Schuch V, Hossack DJ, Chakraborty R, Johnson EL. Melatonin: the placental antioxidant and anti-inflammatory. Front Immunol 2024; 15:1339304. [PMID: 38361952 PMCID: PMC10867115 DOI: 10.3389/fimmu.2024.1339304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Melatonin (N-acetyl-5-methoxytryptamine) is an indolamine hormone with many physiological and biological roles. Melatonin is an antioxidant, anti-inflammatory, free radical scavenger, circadian rhythm regulator, and sleep hormone. However, its most popular role is the ability to regulate sleep through the circadian rhythm. Interestingly, recent studies have shown that melatonin is an important and essential hormone during pregnancy, specifically in the placenta. This is primarily due to the placenta's ability to synthesize its own melatonin rather than depending on the pineal gland. During pregnancy, melatonin acts as an antioxidant and anti-inflammatory, which is necessary to ensure a stable environment for both the mother and the fetus. It is an essential antioxidant in the placenta because it reduces oxidative stress by constantly scavenging for free radicals, i.e., maintain the placenta's integrity. In a healthy pregnancy, the maternal immune system is constantly altered to accommodate the needs of the growing fetus, and melatonin acts as a key anti-inflammatory by regulating immune homeostasis during early and late gestation. This literature review aims to identify and summarize melatonin's role as a powerful antioxidant and anti-inflammatory that reduces oxidative stress and inflammation to maintain a favorable homeostatic environment in the placenta throughout gestation.
Collapse
Affiliation(s)
- Tyana T. Joseph
- Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Viviane Schuch
- Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Daniel J. Hossack
- Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| | - Rana Chakraborty
- Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Erica L. Johnson
- Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, United States
| |
Collapse
|
77
|
Meakin AS, Smith M, Morrison JL, Roberts CT, Lappas M, Ellery SJ, Holland O, Perkins A, McCracken SA, Flenady V, Clifton VL. Placenta-Specific Transcripts Containing Androgen Response Elements Are Altered In Silico by Male Growth Outcomes. Int J Mol Sci 2024; 25:1688. [PMID: 38338965 PMCID: PMC10855055 DOI: 10.3390/ijms25031688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
A birthweight centile (BWC) below the 25th is associated with an elevated risk of adverse perinatal outcomes, particularly among males. This male vulnerability may stem from alterations in placenta-specific androgen signalling, a signalling axis that involves the androgen receptor (AR)-mediated regulation of target genes containing androgen response elements (AREs). In this study, we examined global and ARE-specific transcriptomic signatures in term male placentae (≥37 weeks of gestation) across BWC subcategories (<10th, 10th-30th, >30th) using RNA-seq and gene set enrichment analysis. ARE-containing transcripts in placentae with BWCs below the 10th percentile were upregulated compared to those in the 10th-30th and >30th percentiles, which coincided with the enrichment of gene sets related to hypoxia and the suppression of gene sets associated with mitochondrial function. In the absence of ARE-containing transcripts in silico, <10th and 10th-30th BWC subcategory placentae upregulated gene sets involved in vasculature development, immune function, and cell adhesion when compared to those in the >30th BWC subcategory. Collectively, our in silico findings suggest that changes in the expression of ARE-containing transcripts in male placentae may contribute to impaired placental vasculature and therefore result in reduced fetal growth outcomes.
Collapse
Affiliation(s)
- Ashley S. Meakin
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
- Mater Medical Research Institute, The University of Queensland, Brisbane, QLD 4101, Australia;
| | - Melanie Smith
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (M.S.); (C.T.R.)
| | - Janna L. Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA: Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia;
| | - Claire T. Roberts
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia; (M.S.); (C.T.R.)
| | - Martha Lappas
- Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia;
| | - Stacey J. Ellery
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC 3168, Australia;
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC 3168, Australia
| | - Olivia Holland
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4222, Australia; (O.H.); (A.P.)
| | - Anthony Perkins
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, QLD 4222, Australia; (O.H.); (A.P.)
- School of Health, University of the Sunshine Coast, Sunshine Coast, QLD 4556, Australia
| | - Sharon A. McCracken
- Women and Babies Research, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia;
| | - Vicki Flenady
- Mater Medical Research Institute, The University of Queensland, Brisbane, QLD 4101, Australia;
| | - Vicki L. Clifton
- Mater Medical Research Institute, The University of Queensland, Brisbane, QLD 4101, Australia;
| |
Collapse
|
78
|
Ravelojaona M, Girouard J, Kana Tsapi ES, Chambers M, Vaillancourt C, Van Themsche C, Thornton CA, Reyes-Moreno C. Oncostatin M and STAT3 Signaling Pathways Support Human Trophoblast Differentiation by Inhibiting Inflammatory Stress in Response to IFNγ and GM-CSF. Cells 2024; 13:229. [PMID: 38334621 PMCID: PMC10854549 DOI: 10.3390/cells13030229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/10/2024] Open
Abstract
Interleukin-6 (IL-6) superfamily cytokines play critical roles during human pregnancy by promoting trophoblast differentiation, invasion, and endocrine function, and maintaining embryo immunotolerance and protection. In contrast, the unbalanced activity of pro-inflammatory factors such as interferon gamma (IFNγ) and granulocyte-macrophage colony-stimulating factor (GM-CSF) at the maternal-fetal interface have detrimental effects on trophoblast function and differentiation. This study demonstrates how the IL-6 cytokine family member oncostatin M (OSM) and STAT3 activation regulate trophoblast fusion and endocrine function in response to pro-inflammatory stress induced by IFNγ and GM-CSF. Using human cytotrophoblast-like BeWo (CT/BW) cells, differentiated in villous syncytiotrophoblast (VST/BW) cells, we show that beta-human chorionic gonadotrophin (βhCG) production and cell fusion process are affected in response to IFNγ or GM-CSF. However, those effects are abrogated with OSM by modulating the activation of IFNγ-STAT1 and GM-CSF-STAT5 signaling pathways. OSM stimulation enhances the expression of STAT3, the phosphorylation of STAT3 and SMAD2, and the induction of negative regulators of inflammation (e.g., IL-10 and TGFβ1) and cytokine signaling (e.g., SOCS1 and SOCS3). Using STAT3-deficient VST/BW cells, we show that STAT3 expression is required for OSM to regulate the effects of IFNγ in βhCG and E-cadherin expression. In contrast, OSM retains its modulatory effect on GM-CSF-STAT5 pathway activation even in STAT3-deficient VST/BW cells, suggesting that OSM uses STAT3-dependent and -independent mechanisms to modulate the activation of pro-inflammatory pathways IFNγ-STAT1 and GM-CSF-STAT5. Moreover, STAT3 deficiency in VST/BW cells leads to the production of both a large amount of βhCG and an enhanced expression of activated STAT5 induced by GM-CSF, independently of OSM, suggesting a key role for STAT3 in βhCG production and trophoblast differentiation through STAT5 modulation. In conclusion, our study describes for the first time the critical role played by OSM and STAT3 signaling pathways to preserve and regulate trophoblast biological functions during inflammatory stress.
Collapse
Affiliation(s)
- Marion Ravelojaona
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | - Julie Girouard
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | - Emmanuelle Stella Kana Tsapi
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
| | | | - Cathy Vaillancourt
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
- Centre Armand Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), Laval, QC H7V 1B7, Canada
| | - Céline Van Themsche
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| | | | - Carlos Reyes-Moreno
- Groupe de Recherche en Signalisation Cellulaire (GRSC), Département de Biologie Médicale, Université du Québec à Trois-Rivières, 3351 Boul. des Forges, Trois-Rivières, QC G8Z 4M3, Canada
- Centre de Recherche Interuniversitaire en Reproduction et Développement-Réseau Québécois en Reproduction (CIRD-RQR), Université de Montréal, St-Hyacinthe, QC J2S 2M2, Canada;
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec (RISUQ), Université du Québec, Québec, QC G1K 9H7, Canada
| |
Collapse
|
79
|
Cromb D, Slator P, Hall M, Price A, Alexander D, Counsell S, Hutter J. Advanced magnetic resonance imaging detects altered placental development in pregnancies affected by congenital heart disease. RESEARCH SQUARE 2024:rs.3.rs-3873412. [PMID: 38343847 PMCID: PMC10854304 DOI: 10.21203/rs.3.rs-3873412/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Congenital heart disease (CHD) is the most common congenital malformation and is associated with adverse neurodevelopmental outcomes. The placenta is crucial for healthy fetal development and placental development is altered in pregnancy when the fetus has CHD. This study utilized advanced combined diffusion-relaxation MRI and a data-driven analysis technique to test the hypothesis that placental microstructure and perfusion are altered in CHD-affected pregnancies. 48 participants (36 controls, 12 CHD) underwent 67 MRI scans (50 control, 17 CHD). Significant differences in the weighting of two independent placental and uterine-wall tissue components were identified between the CHD and control groups (both pFDR<0.001), with changes most evident after 30 weeks gestation. A Significant trend over gestation in weighting for a third independent tissue component was also observed in the CHD cohort (R = 0.50, pFDR=0.04), but not in controls. These findings add to existing evidence that placental development is altered in CHD. The results may reflect alterations in placental perfusion or the changes in fetal-placental flow, villous structure and maturation that occur in CHD. Further research is needed to validate and better understand these findings and to understand the relationship between placental development, CHD, and its neurodevelopmental implications.
Collapse
|
80
|
Legault LM, Breton-Larrivée M, Langford-Avelar A, Lemieux A, McGraw S. Sex-based disparities in DNA methylation and gene expression in late-gestation mouse placentas. Biol Sex Differ 2024; 15:2. [PMID: 38183126 PMCID: PMC10770955 DOI: 10.1186/s13293-023-00577-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 12/18/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND The placenta is vital for fetal development and its contributions to various developmental issues, such as pregnancy complications, fetal growth restriction, and maternal exposure, have been extensively studied in mice. The placenta forms mainly from fetal tissue and therefore has the same biological sex as the fetus it supports. Extensive research has delved into the placenta's involvement in pregnancy complications and future offspring development, with a notable emphasis on exploring sex-specific disparities. However, despite these investigations, sex-based disparities in epigenetic (e.g., DNA methylation) and transcriptomic features of the late-gestation mouse placenta remain largely unknown. METHODS We collected male and female mouse placentas at late gestation (E18.5, n = 3/sex) and performed next-generation sequencing to identify genome-wide sex differences in transcription and DNA methylation. RESULTS Our comparison between male and female revealed 358 differentially expressed genes (DEGs) on autosomes, which were associated with signaling pathways involved in transmembrane transport and the responses to viruses and external stimuli. X chromosome DEGs (n = 39) were associated with different pathways, including those regulating chromatin modification and small GTPase-mediated signal transduction. Differentially methylated regions (DMRs) were more common on the X chromosomes (n = 3756) than on autosomes (n = 1705). Interestingly, while most X chromosome DMRs had higher DNA methylation levels in female placentas and tended to be included in CpG dinucleotide-rich regions, 73% of autosomal DMRs had higher methylation levels in male placentas and were distant from CpG-rich regions. Several DEGs were correlated with DMRs. A subset of the DMRs present in late-stage placentas were already established in mid-gestation (E10.5) placentas (n = 348 DMRs on X chromosome and 19 DMRs on autosomes), while others were acquired later in placental development. CONCLUSION Our study provides comprehensive lists of DEGs and DMRs between male and female that collectively cause profound differences in the DNA methylation and gene expression profiles of late-gestation mouse placentas. Our results demonstrate the importance of incorporating sex-specific analyses into epigenetic and transcription studies to enhance the accuracy and comprehensiveness of their conclusions and help address the significant knowledge gap regarding how sex differences influence placental function.
Collapse
Affiliation(s)
- Lisa-Marie Legault
- CHU Ste-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Mélanie Breton-Larrivée
- CHU Ste-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Alexandra Langford-Avelar
- CHU Ste-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC, H3T 1J4, Canada
| | - Anthony Lemieux
- CHU Ste-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Serge McGraw
- CHU Ste-Justine Research Center, 3175 Chemin de La Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.
- Department of Biochemistry and Molecular Medicine, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC, H3T 1J4, Canada.
- Department of Obstetrics and Gynecology, Université de Montréal, 2900 Boulevard Edouard‑Montpetit, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
81
|
Gouvêa VN, Smithyman MM, Hentz F, Bagheri N, Batistel F. Methionine supply during mid-gestation modulates the bovine placental mTOR pathway, nutrient transporters, and offspring birth weight in a sex-specific manner. J Anim Sci 2024; 102:skae305. [PMID: 39390894 DOI: 10.1093/jas/skae305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 10/08/2024] [Indexed: 10/12/2024] Open
Abstract
The mechanistic target of rapamycin (mTOR) predominantly regulates the expression and activity of placental nutrient transporters. The mTOR pathway can be activated by several nutrients, including the essential amino acid methionine. Additionally, previous research in nonruminant animals suggests that mTOR is influenced in a sexually dimorphic manner. In bovine, there is limited understanding of how maternal nutrition and offspring sexual dimorphism affect the placental transfer of nutrients. Thus, we investigated the effects of increasing the supply of dietary methionine to beef cattle heifers during mid-gestation on mTOR signaling, placental nutrient transporters, and fetal growth in male and female offspring. Forty purebred Angus heifers were used in a randomized complete block design experiment. From days 90 to 180 of gestation, heifers received a basal diet with no added methionine (CON, n = 20), or the basal diet plus 8.3 g of rumen-protected methionine (MET, n = 20) per animal daily. All animals received a basal diet in the first and third trimesters of gestation. Cotyledonary tissue samples were collected at parturition and utilized to examine the mTOR pathway and nutrient transporters through protein and gene expression analysis. The offspring's body weight was measured at birth. Data were analyzed using a mixed model that included the fixed effect of treatment, offspring sex, their interactions, and the random effect of block. At day 170 of gestation, MET-supplemented heifers showed higher plasma concentrations of methionine and glutamate (P < 0.01) and lower glycine and proline levels (P ≤ 0.01) compared to the CON group. A treatment × sex interaction was observed for calf birth weight (P = 0.03). In heifers that delivered male calves, MET supplementation increased the birth weight of the calves (P < 0.01). However, the dietary treatments had no effect on the birth weight of female calves (P = 0.32). The increase in birth weight of male calves from MET-fed heifers resembles the upregulation of placental mTOR and phosphorylated mTOR (P ≤ 0.03), as well as the amino acid transporters SLC1A5, SLC7A5, SLC38A6, and SLC38A11, and the glucose transporters SLC2A1 and SLC2A8 (P ≤ 0.05). Our findings suggest that increasing the supply of methionine to beef heifers during mid-gestation can modulate placental nutrient transport and fetal growth in a sex-dependent manner and that these effects are mediated, at least in part, by the mTOR pathway.
Collapse
Affiliation(s)
- Vinícius N Gouvêa
- Texas A&M AgriLife Research and Extension Center, Amarillo, TX 79106, USA
- Department of Animal Science, Texas A&M University, Amarillo, TX 79106, USA
| | - Mackenzie M Smithyman
- Department of Animal and Range Science, Clayton Livestock Research Center, New Mexico State University, Clayton, NM 88415, USA
| | - Fernanda Hentz
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Naghme Bagheri
- Department of Animal, Dairy, and Veterinary Sciences, Utah State University, Logan, UT 84322, USA
| | - Fernanda Batistel
- Department of Animal Sciences, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
82
|
Youyou Z, Zhaoyang L, Chen L, Shuquan Z, Hui W. Effects of Prenatal Methcathinone Exposure on the Neurological Behavior of Adult Offspring. Curr Neuropharmacol 2024; 22:2256-2262. [PMID: 38333971 PMCID: PMC11337688 DOI: 10.2174/1570159x22666240128004722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Our previous research has shown that prenatal methcathinone exposure affects the neurodevelopment and neurobehavior of adolescent offspring, but the study on whether these findings continue into adulthood is limited. OBJECTIVE This study aims to explore the effects of prenatal methcathinone exposure on anxiety-like behavior, learning and memory abilities, as well as serum 5-hydroxytryptamine and dopamine concentrations in adult offspring. METHODS Pregnant rats were injected daily with methcathinone between the 7th and 20th days of gestation. The neurobehavioral performance of both male and female adult offspring rats was evaluated by neurobehavioral tests, including open-field tests, Morris water maze (MWM) tests, and novel object recognition (NOR) tests. The levels of 5-hydroxytryptamine and dopamine concentration in rat serum were detected by ELISA. RESULTS Significant differences were found in the length of center distance and time of center duration in the open-field test, as well as the times of crossing the platform in the MWM test, between the prenatal methcathinone exposure group and the control group. Results of the NOR test showed that adult offspring rats exposed to methcathinone need more time to discriminate the novel object. No gender differences were detected in the neurobehavioral tests. The serum concentrations of 5-hydroxytryptamine and dopamine in rats exposed to methcathinone prenatally were lower than that in the control group, and the serum dopamine concentration was independent of gender in each group. CONCLUSION Prenatal methcathinone exposure affects the neurological behavior in adult offspring, and 5-hydroxytryptamine and dopamine might be involved in the process.
Collapse
Affiliation(s)
- Zhang Youyou
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710004, Shaanxi, P.R. China
- NHC Key Laboratory of Forensic Science, Xi'an Jiaotong University, No. 76, West Yanta Road, Xi'an, 710061, Shaanxi, P.R. China
| | - Li Zhaoyang
- Department of Occupational and Environmental Health, School of Public Health, Xi’an Jiaotong University, No. 76, West Yanta Road, Xi'an, 710061, Shaanxi, P.R. China
| | - Li Chen
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710004, Shaanxi, P.R. China
| | - Zhao Shuquan
- Department of Forensic Pathology, Zhongshan School of Medicine, Sun Yat-sen University, No. 74, Zhongshan 2 Road, Guangzhou, 510080, Guangdong, P.R. China
| | - Wang Hui
- Department of Geriatric Neurology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an Jiaotong University, No. 157, Xiwu Road, Xi'an, 710004, Shaanxi, P.R. China
| |
Collapse
|
83
|
Hattori K, Hoshino Y, Kachi M, Masuda Y, Yamamoto S, Honda S, Minami N, Ikeda S. Analysis of histone H3K4me3 modifications in bovine placenta derived from different calf-production methods. Reprod Domest Anim 2024; 59:e14527. [PMID: 38268203 DOI: 10.1111/rda.14527] [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: 07/01/2023] [Revised: 11/21/2023] [Accepted: 12/28/2023] [Indexed: 01/26/2024]
Abstract
In ruminants, the overgrowth of offspring produced by in vitro fertilization (IVF) is a common problem. Abnormal epigenetic modifications caused by environmental factors during the early embryonic period are suspected as an aetiology of overgrowth. In this study, we investigated the genome-wide histone H3K4me3 profiles of bovine placentae that play a pivotal role in foetal development and compared their characteristics between artificial insemination (AI)- and IVF-derived samples. Cotyledons were harvested from the placentae obtained at parturition of 5 AI- and 13 IVF-derived calves, and chromatin immunoprecipitation sequencing was performed for H3K4me3. We confirmed no significant maternal tissue contamination in the samples we used. The revealed H3K4me3 profiles reflected the general characteristics of the H3K4me3 modification, which is abundantly distributed in the promoter region of active genes. By extracting common modifications from multiple samples, the genes involved in placenta-specific biological processes could be enriched. Comparison with the H3K4me3 modifications of blastocyst samples was also effective for enriching the placenta-specific features. Principal component analysis suggested the presence of differential H3K4me3 modifications in AI- and IVF-derived samples. The genes contributing to the difference were related to the developmental biological processes. Imprinted genes such as BEGAIN, ZNF215 and DLX5 were among the extracted genes. Principal component and discriminant analyses using only male samples categorized the samples into three groups based on foetal weight and calf-production methods. To our knowledge, this is the first study to profile the genome-wide histone modifications of bovine foetal placentae and reveal their differential characteristics between different calf-production methods.
Collapse
Affiliation(s)
- Kanoko Hattori
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Yoichiro Hoshino
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masayuki Kachi
- Dairy Research Department, Gifu Prefectural Livestock Research Institute, Ena, Gifu, Japan
| | - Yasumitsu Masuda
- Department of Animal Science, Tottori Livestock Research Center, Tottori, Japan
| | - Satoshi Yamamoto
- Livestock Technology Research Center, Hiroshima Prefectural Technology Research Institute, Shobara, Hiroshima, Japan
| | - Shinnosuke Honda
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Naojiro Minami
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Shuntaro Ikeda
- Laboratory of Reproductive Biology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| |
Collapse
|
84
|
Manasova GS, Stasy YA, Kaminsky VV, Gladchuk IZ, Nitochko EA. Histological and immunohistochemical features of the placenta associated with COVID-19: a systematic review and meta-analysis. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2024; 77:1434-1455. [PMID: 39241144 DOI: 10.36740/wlek202407120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2024]
Abstract
OBJECTIVE Aim: To make a systematic review and meta-analysis of published data on the study of histological and immunohistochemical features of the placenta in women who had acute coronavirus infection associated with SARS-CoV-2 ("Covid" placentas) during pregnancy. PATIENTS AND METHODS Materials and Methods: The search for literature data is based on the PRISMA methodology); the MEDLINE database (PubMed®) was searched using Medical Subject Headings terms from January 2020 to July 2023. The project was registered in the Open Sience Frame (Project Identifier: DOI 10.17605/OSF.IO/GDR3S, Registration DOI: https://doi.org/10.17605/OSF.IO/H2KPU). Preference was given to studies in which the description of placentas met the requirements of the Amsterdam Placental Workshop Group Consensus Statement. RESULTS Results: A total of 31 studies were included; the number of participants whose morphological and histological description of the placentas could be subjected to meta-analysis was 2401, respectively, in the group with a "Covid" history and 1910 - conditionally healthy pregnant women. Pathological changes in the placental complex were not detected in 42±19.62% of pregnant women with a history of Covid. Immunohistochemical examination of placentas preferably focuses on the detection of SARS-CoV-2 spike protein or ACE2. According to currently available studies, in the placentas of women who have had COVID-19 during pregnancy, there are no pathognomic histological patterns specific to this infection and direct damage to the placenta is rarely observed. Histological patterns in "covid" placentas are isolated, most often a combination of lesions in both the maternal and fetal malperfusion. CONCLUSION Conclusions: According to currently available studies, in the placentas of women who have had COVID-19 during pregnancy, there are no pathognomic histological patterns specific to this infection and direct damage to the placenta is rarely observed. The probability of infection of the intrauterine fetus by the transplacental hematogenous route is the lowest compared to other routes, which, in our opinion, is a possible explanation for the high frequency of MVM without subsequent infection of the fetus.
Collapse
Affiliation(s)
| | - Yana A Stasy
- ODESA NATIONAL MEDICAL UNIVERSITY, ODESA, UKRAINE
| | | | | | | |
Collapse
|
85
|
Zhang Y, Yang H. Quantification of Trophoblast Syncytialization by Fluorescent Membrane Labeling. Methods Mol Biol 2024; 2728:99-104. [PMID: 38019394 DOI: 10.1007/978-1-0716-3495-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Trophoblast fusion or syncytialization is a fundamental yet poorly understood process during placental development. Primary cultured cytotrophoblasts and human choriocarcinoma cell lines are commonly used to study trophoblast fusion mechanisms in vitro. Quantification of trophoblast fusion index is a key for the in vitro studies. In this chapter, we describe a new method to quantify fusion index, which is based on fluorescent labeling of the plasma membrane using Di-8-ANEPPS, a membrane potential dye. This method directly works on live cells, thereby is simple, economic, and reliable.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA.
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
86
|
Díaz L, Olmos-Ortiz A, Flores-Espinosa P, Mancilla-Herrera I, Zaga-Clavellina V. In Vitro Culturing of Human Trophoblasts from Term Placenta. Methods Mol Biol 2024; 2781:47-59. [PMID: 38502442 DOI: 10.1007/978-1-0716-3746-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Since the early 1960s, researchers began culturing placental cells to establish an in vitro model to study the biology of human trophoblasts, including their ability to differentiate into syncytiotrophoblasts and secrete steroid and peptide hormones that help sustain a viable pregnancy. This task was addressed by testing different serum concentrations, cell culture media, digestive enzymes, growth factors, substrate coating with diverse proteins from the extracellular matrix, and so on. Among the many methodological challenges, the contamination of trophoblasts with other cell types, such as immune and stromal cells, was a matter of concern. However, introducing the Percoll gradient to isolate cytotrophoblasts was an excellent contribution, and later, the depletion of contaminating cells by using magnetic bead-conjugated antibodies also helped increase the purity of cytotrophoblasts. Herein, with some modifications, we describe a rapid and easy method for cytotrophoblast isolation from the term human placenta based on the previously reported method by Harvey Kliman et al. (Endocrinology 118:1567-1582, 1986). This method yields about 40-90 million cells from a single placenta, with a purity of around 85-90%.
Collapse
Affiliation(s)
- Lorenza Díaz
- Departamento de Biología de la Reproducción Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Andrea Olmos-Ortiz
- Department of Immunobiochemistry Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de Mexico, Mexico
| | - Pilar Flores-Espinosa
- Department of Immunobiochemistry Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de Mexico, Mexico
| | - Ismael Mancilla-Herrera
- Department of Infectology and Immunology, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | - Verónica Zaga-Clavellina
- Department of Immunobiochemistry Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Ciudad de Mexico, Mexico
| |
Collapse
|
87
|
Meng Q, Del Rosario I, Sung K, Janzen C, Devaskar SU, Carpenter CL, Ritz B. Maternal dietary patterns and placental outcomes among pregnant women in Los Angeles. Placenta 2024; 145:72-79. [PMID: 38100961 PMCID: PMC11419549 DOI: 10.1016/j.placenta.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/11/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Epidemiological studies have linked prenatal maternal diet to fetal growth, but whether diet affects placental outcomes is poorly understood. METHODS We collected past month dietary intake from 148 women in mid-pregnancy enrolled at University of California Los Angeles (UCLA) antenatal clinics from 2016 to 2019. We employed the food frequency Diet History Questionnaire II and generated the Healthy Eating Index-2015 (HEI-2015), the Alternate Healthy Eating Index for Pregnancy (AHEI-P), and the Alternate Mediterranean Diet (aMED). We conducted T2-weighted magnetic resonance imaging (MRI) in mid-pregnancy (1st during 14-17 and 2nd during 19-24 gestational weeks) to evaluate placental volume (cm3) and we measured placenta weight (g) at delivery. We estimated change and 95 % confidence interval (CI) in placental volume and associations of placenta weight with all dietary index scores and diet items using linear regression models. RESULTS Placental volume in mid-pregnancy was associated with an 18.9 cm3 (95 % CI 5.1, 32.8) increase per 100 gestational days in women with a higher HEI-2015 (≥median), with stronger results for placentas of male fetuses. We estimated positive associations between placental volume at the 1st and 2nd MRI and higher intake of vegetables, high-fat fish, dairy, and dietary intake of B vitamins. A higher aMED (≥median) score was associated with a 40.5 g (95 % CI 8.5, 72.5) increase in placenta weight at delivery, which was mainly related to protein intake. DISCUSSION Placental growth represented by volume in mid-pregnancy and weight at birth is influenced by the quality and content of the maternal diet.
Collapse
Affiliation(s)
- Qi Meng
- Department of Epidemiology, University of California, Los Angeles, CA, 90095, USA
| | - Irish Del Rosario
- Department of Epidemiology, University of California, Los Angeles, CA, 90095, USA
| | - Kyunghyun Sung
- Department of Radiological Sciences, University of California, Los Angeles, CA, 90095, USA
| | - Carla Janzen
- Department of Obstetrics & Gynecology, University of California, Los Angeles, CA, 90095, USA
| | - Sherin U Devaskar
- Department of Pediatrics, University of California, Los Angeles, CA, 90095, USA
| | | | - Beate Ritz
- Department of Epidemiology, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
88
|
Abubaker S, Miri S, Mottawea W, Hammami R. Microbial Extracellular Vesicles in Host-Microbiota Interactions. Results Probl Cell Differ 2024; 73:475-520. [PMID: 39242390 DOI: 10.1007/978-3-031-62036-2_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2024]
Abstract
Extracellular vesicles have emerged as key players in cellular communication, influencing various physiological processes and pathophysiological progression, including digestion, immune response, and tissue repairs. Recently, a class of EVs derived from microbial communities has gained significant attention due to their pivotal role in intercellular communication and their potential as biomarkers and biotherapeutic agents. Microbial EVs are membrane-bound molecules encapsulating bioactive metabolites that modulate host physiological and pathological processes. This chapter discusses the evolving history of microbiota-produced EVs, including their discovery, characterization, current research status, and their diverse mechanisms of interaction with other microbes and hosts. This review also highlights the importance of EVs in health and disease and discusses recent research that shows promising results for the therapeutic potential of EVs.
Collapse
Affiliation(s)
- Sarah Abubaker
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Saba Miri
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Walid Mottawea
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Riadh Hammami
- Faculty of Health Sciences, School of Nutrition Sciences, University of Ottawa, Ottawa, ON, Canada.
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
89
|
Islam M, Behura SK. Role of paralogs in the sex-bias transcriptional and metabolic regulation of the brain-placental axis in mice. Placenta 2024; 145:143-150. [PMID: 38134547 DOI: 10.1016/j.placenta.2023.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
INTRODUCTION Duplicated genes or paralogs play important roles in the adaptive function of eukaryotic genomes. Animal studies have shown evidence for the functional role of paralogs in pregnancy, but our knowledge about the role of paralogs in the fetoplacental regulation remains limited. In particular, if fetoplacental metabolic regulation is modulated by differential expression of paralogs remains unexamined. METHODS In this study, gene expression profiles of day-15 placenta and fetal brain were compared to identify families or groups of paralogous genes expressed in the placenta and brain of male versus female fetuses in mice. A Bayesian modeling was applied to infer directional relationship of transcriptional variation of the paralogs relative to the phylogenetic variation of the genes in each family. Gas chromatography-mass spectrometry (GC-MS) was used to perform untargeted metabolomics analysis of day-15 placenta and fetal brain of both sexes. RESULTS We identified paralog groups that were expressed in a sex and/or tissue biased manner between the placenta and fetal brain. Bayesian modeling showed evidence for directional relationship between expression and phylogeny of specific paralogs. These relationships were sex specific. GC-MS analysis identified metabolites that were expressed in a sex-bias manner between the placenta and fetal brain. By performing integrative analysis of the metabolomics and gene expression data, we showed that specific groups of metabolites and paralogous genes were expressed in a coordinated manner between the placenta and fetal brain. DISCUSSION The findings of this study collectively suggest that paralogs play an influential role in the regulation of the brain-placental axis in mice.
Collapse
Affiliation(s)
- Maliha Islam
- Division of Animal Sciences, University of Missouri, 920 East Campus Drive, Columbia, Missouri, 65211, USA
| | - Susanta K Behura
- Division of Animal Sciences, University of Missouri, 920 East Campus Drive, Columbia, Missouri, 65211, USA; MU Institute for Data Science and Informatics, University of Missouri, USA; Interdisciplinary Reproduction and Health Group, University of Missouri, USA; Interdisciplinary Neuroscience Program, University of Missouri, USA.
| |
Collapse
|
90
|
Chang Y, Zhou Y, Zhou J, Li W, Cao J, Jing Y, Zhang S, Shen Y, Lin Q, Fan X, Yang H, Dong X, Zhang S, Yi X, Shuai L, Shi L, Liu Z, Yang J, Ma X, Hao J, Chen K, Li MJ, Wang F, Huang D. Unraveling the causal genes and transcriptomic determinants of human telomere length. Nat Commun 2023; 14:8517. [PMID: 38129441 PMCID: PMC10739845 DOI: 10.1038/s41467-023-44355-z] [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: 01/18/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Telomere length (TL) shortening is a pivotal indicator of biological aging and is associated with many human diseases. The genetic determinates of human TL have been widely investigated, however, most existing studies were conducted based on adult tissues which are heavily influenced by lifetime exposure. Based on the analyses of terminal restriction fragment (TRF) length of telomere, individual genotypes, and gene expressions on 166 healthy placental tissues, we systematically interrogate TL-modulated genes and their potential functions. We discover that the TL in the placenta is comparatively longer than in other adult tissues, but exhibiting an intra-tissue homogeneity. Trans-ancestral TL genome-wide association studies (GWASs) on 644,553 individuals identify 20 newly discovered genetic associations and provide increased polygenic determination of human TL. Next, we integrate the powerful TL GWAS with placental expression quantitative trait locus (eQTL) mapping to prioritize 23 likely causal genes, among which 4 are functionally validated, including MMUT, RRM1, KIAA1429, and YWHAZ. Finally, modeling transcriptomic signatures and TRF-based TL improve the prediction performance of human TL. This study deepens our understanding of causal genes and transcriptomic determinants of human TL, promoting the mechanistic research on fine-grained TL regulation.
Collapse
Affiliation(s)
- Ying Chang
- Tianjin Key Lab of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University, Tianjin, China
| | - Yao Zhou
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Junrui Zhou
- Department of Genetics and Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Wen Li
- Tianjin Key Lab of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University, Tianjin, China
| | - Jiasong Cao
- Tianjin Key Lab of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University, Tianjin, China
| | - Yaqing Jing
- Department of Genetics and Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shan Zhang
- Department of Genetics and Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yongmei Shen
- Tianjin Key Lab of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University, Tianjin, China
| | - Qimei Lin
- Tianjin Key Lab of Human Development and Reproductive Regulation, Tianjin Central Hospital of Obstetrics and Gynecology, Nankai University, Tianjin, China
| | - Xutong Fan
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hongxi Yang
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaobao Dong
- Department of Genetics and Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Shijie Zhang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xianfu Yi
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Ling Shuai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Tianjin Central Hospital of Gynecology Obstetrics/Tianjin Key Laboratory of Human Development and Reproductive Regulation, Nankai University, Tianjin, China
| | - Lei Shi
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhe Liu
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jie Yang
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Jihui Hao
- Department of Pancreatic Cancer, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Epidemiology and Biostatistics, Tianjin Key Laboratory of Molecular Cancer Epidemiology, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| | - Feng Wang
- Department of Genetics and Tianjin Key Laboratory of Cellular and Molecular Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Tianjin Medical University School of Stomatology, Tianjin Medical University, Tianjin, China.
- Department of Geriatrics, Tianjin Medical University General Hospital; Tianjin Geriatrics Institute, Tianjin, China.
| | - Dandan Huang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
91
|
Gorman-Sandler E, Wood G, Cloude N, Frambes N, Brennen H, Robertson B, Hollis F. Mitochondrial might: powering the peripartum for risk and resilience. Front Behav Neurosci 2023; 17:1286811. [PMID: 38187925 PMCID: PMC10767224 DOI: 10.3389/fnbeh.2023.1286811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/01/2023] [Indexed: 01/09/2024] Open
Abstract
The peripartum period, characterized by dynamic hormonal shifts and physiological adaptations, has been recognized as a potentially vulnerable period for the development of mood disorders such as postpartum depression (PPD). Stress is a well-established risk factor for developing PPD and is known to modulate mitochondrial function. While primarily known for their role in energy production, mitochondria also influence processes such as stress regulation, steroid hormone synthesis, glucocorticoid response, GABA metabolism, and immune modulation - all of which are crucial for healthy pregnancy and relevant to PPD pathology. While mitochondrial function has been implicated in other psychiatric illnesses, its role in peripartum stress and mental health remains largely unexplored, especially in relation to the brain. In this review, we first provide an overview of mitochondrial involvement in processes implicated in peripartum mood disorders, underscoring their potential role in mediating pathology. We then discuss clinical and preclinical studies of mitochondria in the context of peripartum stress and mental health, emphasizing the need for better understanding of this relationship. Finally, we propose mitochondria as biological mediators of resilience to peripartum mood disorders.
Collapse
Affiliation(s)
- Erin Gorman-Sandler
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Columbia VA Healthcare System, Columbia, SC, United States
| | - Gabrielle Wood
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Nazharee Cloude
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Noelle Frambes
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Hannah Brennen
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Breanna Robertson
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Fiona Hollis
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, United States
- Columbia VA Healthcare System, Columbia, SC, United States
- USC Institute for Cardiovascular Disease Research, Columbia, SC, United States
| |
Collapse
|
92
|
Batorsky R, Ceasrine AM, Shook LL, Kislal S, Bordt EA, Devlin BA, Perlis RH, Slonim DK, Bilbo SD, Edlow AG. Hofbauer cells and fetal brain microglia share transcriptional profiles and responses to maternal diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.16.571680. [PMID: 38187648 PMCID: PMC10769274 DOI: 10.1101/2023.12.16.571680] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Maternal immune activation is associated with adverse offspring neurodevelopmental outcomes, many mediated by in utero microglial programming. As microglia remain inaccessible throughout development, identification of noninvasive biomarkers reflecting fetal brain microglial programming could permit screening and intervention. We used lineage tracing to demonstrate the shared ontogeny between fetal brain macrophages (microglia) and fetal placental macrophages (Hofbauer cells) in a mouse model of maternal diet-induced obesity, and single-cell RNA-seq to demonstrate shared transcriptional programs. Comparison with human datasets demonstrated conservation of placental resident macrophage signatures between mice and humans. Single-cell RNA-seq identified common alterations in fetal microglial and Hofbauer cell gene expression induced by maternal obesity, as well as sex differences in these alterations. We propose that Hofbauer cells, which are easily accessible at birth, provide novel insights into fetal brain microglial programs, and may facilitate the early identification of offspring vulnerable to neurodevelopmental disorders in the setting of maternal exposures.
Collapse
Affiliation(s)
| | - Alexis M. Ceasrine
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Lydia L. Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evan A. Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Benjamin A. Devlin
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
| | - Roy H. Perlis
- Department of Psychiatry and Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Donna K. Slonim
- Department of Computer Science, Tufts University, Medford, MA
| | - Staci D. Bilbo
- Department of Psychology and Neuroscience, Duke University, Durham, NC, USA
- Department of Neurobiology, Duke University, Durham, NC, USA
- Lurie Center for Autism, Massachusetts General Hospital, Boston, MA
| | - Andrea G. Edlow
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
93
|
Toudic C, Maurer M, St-Pierre G, Xiao Y, Bannert N, Lafond J, Rassart É, Sato S, Barbeau B. Galectin-1 Modulates the Fusogenic Activity of Placental Endogenous Retroviral Envelopes. Viruses 2023; 15:2441. [PMID: 38140682 PMCID: PMC10747188 DOI: 10.3390/v15122441] [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: 10/24/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Syncytin-1 and -2 are glycoproteins encoded by human endogenous retrovirus (hERV) that, through their fusogenic properties, are needed for the formation of the placental syncytiotrophoblast. Previous studies suggested that these proteins, in addition to the EnvP(b) envelope protein, are also involved in other cell fusion events. Since galectin-1 is a β-galactoside-binding protein associated with cytotrophoblast fusion during placental development, we previously tested its effect on Syncytin-mediated cell fusion and showed that this protein differently modulates the fusogenic potential of Syncytin-1 and -2. Herein, we were interested in comparing the impact of galectin-1 on hERV envelope proteins in different cellular contexts. Using a syncytium assay, we first demonstrated that galectin-1 increased the fusion of Syncytin-2- and EnvP(b)-expressing cells. We then tested the infectivity of Syncytin-1 and -2 vs. VSV-G-pseudotyped viruses toward Cos-7 and various human cell lines. In the presence of galectin-1, infection of Syncytin-2-pseudotyped viruses augmented for all cell lines. In contrast, the impact of galectin-1 on the infectivity of Syncytin-1-pseudotyped viruses varied, being cell- and dose-dependent. In this study, we report the functional associations between three hERV envelope proteins and galectin-1, which should provide information on the fusogenic activity of these proteins in the placenta and other biological and pathological processes.
Collapse
Affiliation(s)
- Caroline Toudic
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
| | - Maike Maurer
- Robert-Koch Institute, 13353 Berlin, Germany; (M.M.); (N.B.)
| | - Guillaume St-Pierre
- Glycobiology and Bioimaging Laboratory, Research Centre for Infectious Diseases and Axe Maladies Infectieuses et Immunitaires, Laval University, Quebec City, QC G1V 0A6, Canada; (G.S.-P.); (S.S.)
| | - Yong Xiao
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
| | - Norbert Bannert
- Robert-Koch Institute, 13353 Berlin, Germany; (M.M.); (N.B.)
| | - Julie Lafond
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
| | - Éric Rassart
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
| | - Sachiko Sato
- Glycobiology and Bioimaging Laboratory, Research Centre for Infectious Diseases and Axe Maladies Infectieuses et Immunitaires, Laval University, Quebec City, QC G1V 0A6, Canada; (G.S.-P.); (S.S.)
| | - Benoit Barbeau
- Département des Sciences Biologiques and Centre d’excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Université du Québec à Montréal, Montréal, QC H3C 3P8, Canada; (C.T.); (Y.X.); (J.L.); (É.R.)
- Regroupement Intersectoriel de Recherche en Santé de l’Université du Québec, Montréal, QC H2X 1E3, Canada
| |
Collapse
|
94
|
Fives C, Toulouse A, Kenny L, Brosnan T, McCarthy J, Fitzgerald B. Cytology Techniques Can Provide Insight into Human Placental Structure Including Syncytiotrophoblast Nuclear Spatial Organisation. J Dev Biol 2023; 11:46. [PMID: 38132714 PMCID: PMC10743966 DOI: 10.3390/jdb11040046] [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: 11/17/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
The aim of this study was to provide the first systematic description of human placental cytology appearances and to investigate syncytiotrophoblast nuclear organisation patterns using cytology techniques. Term placentas from normal pregnancies were sampled using fine-needle aspiration (FNA) and direct scrapes. Standard histological examination was also performed to exclude pathological changes in the placentas being studied. Both Papanicolaou-stained cytospin preparations and air-dried Giemsa slides from FNA provided high-quality material for cytological assessment with good cellularity. Among the key features of the cytology preparations were villous "microbiopsies" that allowed for the three-dimensional appreciation of villous branching patterns. Cytological appearances, including nuclear characteristics of villous cytotrophoblast and syncytiotrophoblast, were also well demonstrated. In microbiopsies and detached villous trophoblast sheets, complex patterns of syncytiotrophoblast nuclear organisation, not previously described cytologically, were observed, including irregular spacing of nuclei, syncytioplasm windows and linear nuclear arrangements. This study showed that placental cytology (a) provides technically excellent material for cytological evaluation, (b) confirms the presence of complex nuclear organisational patterns in the syncytiotrophoblast by eliminating the possibility of tangential sectioning artefact, (c) provides superior nuclear detail over standard histological sections and (d) may be an untapped research resource for the investigation of normal and pathological processes because of its ability to look at the placenta in a novel way and through its potential for both ex vivo and in vivo placental sampling.
Collapse
Affiliation(s)
- Cassie Fives
- Department of Pathology, Cork University Hospital, T12 DC4A Cork, Ireland
| | - André Toulouse
- Department of Anatomy and Neuroscience, University College Cork, T12 XF62 Cork, Ireland
| | - Louise Kenny
- Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7TX, UK
| | - Therese Brosnan
- Department of Pathology, Cork University Hospital, T12 DC4A Cork, Ireland
- Pregnancy Loss Research Group, Department of Obstetrics and Gynaecology, University College Cork, T12 YE02 Cork, Ireland
| | - Julie McCarthy
- Department of Pathology, Cork University Hospital, T12 DC4A Cork, Ireland
| | - Brendan Fitzgerald
- Department of Pathology, Cork University Hospital, T12 DC4A Cork, Ireland
- Pregnancy Loss Research Group, Department of Obstetrics and Gynaecology, University College Cork, T12 YE02 Cork, Ireland
| |
Collapse
|
95
|
Rogers LM, Huggins M, Doster RS, Omage JI, Gaddy JA, Eastman A, Aronoff DM. Impact of Metabolic Stress on BeWo Syncytiotrophoblast Function. Chembiochem 2023; 24:e202300410. [PMID: 37800606 DOI: 10.1002/cbic.202300410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/07/2023]
Abstract
During placental formation, cytotrophoblasts (CTBs) fuse into multinucleate, microvilli-coated syncytiotrophoblasts (STBs), which contact maternal blood, mediating nutrient, metabolite, and gas exchange between mother and fetus, and providing a barrier against fetal infection. Trophoblasts remodel the surrounding extracellular matrix through the secretion of matrix metalloproteinases (MMPs). Maternal obesity and diabetes mellitus can negatively impact fetal development and may impair trophoblast function. We sought to model the impact of metabolic stress on STB function by examining MMP and hormone secretion. The BeWo CTB cell line was syncytialized to STB-like cells with forskolin. Cell morphology was examined by electron microscopy and immunofluorescence; phenotype was further assessed by ELISA and RT-qPCR. STBs were exposed to a metabolic stress cocktail (MetaC: 30 mM glucose, 10 nM insulin, and 0.1 mM palmitic acid). BeWo syncytialization was demonstrated by increased secretion of HCGβ and progesterone, elevated syncytin gene expression (ERVW-1 and ERVFRD-1), loss of tight junctions, and increased surface microvilli. MetaC strongly suppressed syncytin gene expression (ERVW-1 and ERVFRD-1), suppressed HCGβ and progesterone secretion, and altered both MMP-9 and MMP-2 production. Metabolic stress modeling diabetes and obesity altered BeWo STB hormone and MMP production in vitro.
Collapse
Affiliation(s)
- Lisa M Rogers
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, 545 Barnhill Dr., EH 305, Indianapolis, IN, 46202, USA
| | - Marissa Huggins
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Ryan S Doster
- Division of Infectious Diseases, Department of Medicine, Department of Microbiology and Immunology, University of Louisville, Louisville, KY, 40202, USA
| | - Joel I Omage
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Jennifer A Gaddy
- Division of Infectious Diseases, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Alison Eastman
- Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - David M Aronoff
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, 545 Barnhill Dr., EH 305, Indianapolis, IN, 46202, USA
| |
Collapse
|
96
|
Owen DM, Kwon M, Huang X, Nagari A, Nandu T, Kraus WL. Genome-wide identification of transcriptional enhancers during human placental development and association with function, differentiation, and disease†. Biol Reprod 2023; 109:965-981. [PMID: 37694817 PMCID: PMC10724456 DOI: 10.1093/biolre/ioad119] [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: 01/28/2023] [Revised: 08/07/2023] [Accepted: 09/09/2023] [Indexed: 09/12/2023] Open
Abstract
The placenta is a dynamic organ that must perform a remarkable variety of functions during its relatively short existence in order to support a developing fetus. These functions include nutrient delivery, gas exchange, waste removal, hormone production, and immune barrier protection. Proper placenta development and function are critical for healthy pregnancy outcomes, but the underlying genomic regulatory events that control this process remain largely unknown. We hypothesized that mapping sites of transcriptional enhancer activity and associated changes in gene expression across gestation in human placenta tissue would identify genomic loci and predicted transcription factor activity related to critical placenta functions. We used a suite of genomic assays [i.e., RNA-sequencing (RNA-seq), Precision run-on-sequencing (PRO-seq), and Chromatin immunoprecipitation-sequencing (ChIP-seq)] and computational pipelines to identify a set of >20 000 enhancers that are active at various time points in gestation. Changes in the activity of these enhancers correlate with changes in gene expression. In addition, some of these enhancers encode risk for adverse pregnancy outcomes. We further show that integrating enhancer activity, transcription factor motif analysis, and transcription factor expression can identify distinct sets of transcription factors predicted to be more active either in early pregnancy or at term. Knockdown of selected identified transcription factors in a trophoblast stem cell culture model altered the expression of key placental marker genes. These observations provide a framework for future mechanistic studies of individual enhancer-transcription factor-target gene interactions and have the potential to inform genetic risk prediction for adverse pregnancy outcomes.
Collapse
Affiliation(s)
- David M Owen
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of General Obstetrics and Gynecology, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Minjung Kwon
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuan Huang
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anusha Nagari
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tulip Nandu
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - W Lee Kraus
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Basic Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
97
|
Ashley RL, Trigo EM, Ervin JM. Placental insufficiency and heavier placentas in sheep after suppressing CXCL12/CXCR4 signaling during implantation†. Biol Reprod 2023; 109:982-993. [PMID: 37724932 PMCID: PMC10724462 DOI: 10.1093/biolre/ioad122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/28/2023] [Accepted: 09/15/2023] [Indexed: 09/21/2023] Open
Abstract
During implantation, trophoblast cell invasion and differentiation is predominantly important to achieving proper placental formation and embryonic development. The chemokine, C-X-C motif chemokine ligand 12 (CXCL12) working through its receptor C-X-C motif chemokine receptor 4 (CXCR4) is implicated in implantation and placentation but precise roles of this axis are unclear. Suppressing CXCL12/CXCR4 signaling at the fetal-maternal interface in sheep reduces trophoblast invasion, disrupts uterine remodeling, and diminishes placental vascularization. We hypothesize these negative impacts during implantation will manifest as compromised fetal and placental growth at midgestation. To test, on day 12 postbreeding, osmotic pumps were surgically installed in 30 ewes and delivered intrauterine CXCR4 inhibitor or saline for 7 or 14 days. On day 90, fetal/maternal tissues were collected, measured, weighed, and maternal (caruncle) and fetal (cotyledon) placenta components separated and analyzed. The objectives were to determine if (i) suppressing CXCL12/CXCR4 during implantation results in reduced fetal and placental growth and development and (ii) if varying the amount of time CXCL12/CXCR4 is suppressed impacts fetal/placental development. Fetal weights were similar; however greater placental weight and placentome numbers occurred when CXCL12/CXCR4 was suppressed for 14 days. In caruncles, greater abundance of fibroblast growth factor 2, vascular endothelial growth factor A, vascular endothelial growth factor A receptor 1 (FLT-1), and placental growth factor were observed after suppressing CXCL12/CXCR4. Similar results occurred in cotyledons except less vascular endothelial growth factor in 7 day group and less fibroblast growth factor in 14 day group. Our data underscore the importance of CXCL12/CXCR4 signaling during placentation and provide strong evidence that altering CXCL12-mediated signaling induces enduring placental effects manifesting later in gestation.
Collapse
Affiliation(s)
- Ryan L Ashley
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| | - Elisa M Trigo
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| | - Jacqueline M Ervin
- Department of Animal and Range Sciences, New Mexico State University, Las Cruces, NM, USA
| |
Collapse
|
98
|
Elkin ER, Campbell KA, Lapehn S, Harris SM, Padmanabhan V, Bakulski KM, Paquette AG. Placental single cell transcriptomics: Opportunities for endocrine disrupting chemical toxicology. Mol Cell Endocrinol 2023; 578:112066. [PMID: 37690473 PMCID: PMC10591899 DOI: 10.1016/j.mce.2023.112066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/12/2023]
Abstract
The placenta performs essential biologic functions for fetal development throughout pregnancy. Placental dysfunction is at the root of multiple adverse birth outcomes such as intrauterine growth restriction, preeclampsia, and preterm birth. Exposure to endocrine disrupting chemicals during pregnancy can cause placental dysfunction, and many prior human studies have examined molecular changes in bulk placental tissues. Placenta-specific cell types, including cytotrophoblasts, syncytiotrophoblasts, extravillous trophoblasts, and placental resident macrophage Hofbauer cells play unique roles in placental development, structure, and function. Toxicant-induced changes in relative abundance and/or impairment of these cell types likely contribute to placental pathogenesis. Although gene expression insights gained from bulk placental tissue RNA-sequencing data are useful, their interpretation is limited because bulk analysis can mask the effects of a chemical on individual populations of placental cells. Cutting-edge single cell RNA-sequencing technologies are enabling the investigation of placental cell-type specific responses to endocrine disrupting chemicals. Moreover, in situ bioinformatic cell deconvolution enables the estimation of cell type proportions in bulk placental tissue gene expression data. These emerging technologies have tremendous potential to provide novel mechanistic insights in a complex heterogeneous tissue with implications for toxicant contributions to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Elana R Elkin
- School of Public Health, San Diego State University, San Diego, CA, USA.
| | - Kyle A Campbell
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Samantha Lapehn
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA
| | - Sean M Harris
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, Michigan Medicine, Ann Arbor, MI, USA
| | - Kelly M Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
99
|
Kouthouridis S, Sotra A, Khan Z, Alvarado J, Raha S, Zhang B. Modeling the Progression of Placental Transport from Early- to Late-Stage Pregnancy by Tuning Trophoblast Differentiation and Vascularization. Adv Healthc Mater 2023; 12:e2301428. [PMID: 37830445 PMCID: PMC11468690 DOI: 10.1002/adhm.202301428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/18/2023] [Indexed: 10/14/2023]
Abstract
The early-stage placental barrier is characterized by a lack of fetal circulation and by a thick trophoblastic barrier, whereas the later-stage placenta consists of vascularized chorionic villi encased in a thin, differentiated trophoblast layer, ideal for nutrient transport. In this work, predictive models of early- and late-stage placental transport are created using blastocyst-derived placental stem cells (PSCs) by modulating PSC differentiation and model vascularization. PSC differentiation results in a thinner, fused trophoblast layer, as well as an increase in human chorionic gonadotropin secretion, barrier permeability, and secretion of certain inflammatory cytokines, which are consistent with in vivo findings. Further, gene expression confirms this shift toward a differentiated trophoblast subtype. Vascularization results in a molecule type- and size-dependent change in dextran and insulin permeability. These results demonstrate that trophoblast differentiation and vascularization have critical effects on placental barrier permeability and that this model can be used as a predictive measure to assess fetal toxicity of xenobiotic substances at different stages of pregnancy.
Collapse
Affiliation(s)
- Sonya Kouthouridis
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Alexander Sotra
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Zaim Khan
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Justin Alvarado
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Sandeep Raha
- Department of Pediatrics and the Graduate Programme in Medical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Boyang Zhang
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| |
Collapse
|
100
|
Adams S, Stapleton PA. Nanoparticles at the maternal-fetal interface. Mol Cell Endocrinol 2023; 578:112067. [PMID: 37689342 PMCID: PMC10591848 DOI: 10.1016/j.mce.2023.112067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/25/2023] [Accepted: 09/05/2023] [Indexed: 09/11/2023]
Abstract
The increasing production of intentional and unintentional nanoparticles (NPs) has led to their accumulation in the environment as air and ground pollution. The heterogeneity of these particles primarily relies on the NP physicochemical properties (i.e., chemical composition, size, shape, surface chemistry, etc.). Pregnancy represents a vulnerable life stage for both the woman and the developing fetus. The ubiquitous nature of these NPs creates a concern for developmental fetal exposures. At the maternal-fetal interface lies the placenta, a temporary endocrine organ that facilitates nutrient and waste exchange as well as communication between maternal and fetal tissues. Recent evidence in human and animal models identifies that gestational exposure to NPs results in placental translocation leading to local effects and endocrine disruption. Currently, the mechanisms underlying placental translocation and cellular uptake of NPs in the placenta are poorly understood. The purpose of this review is to assess the current understanding of the physiochemical factors influencing NP translocation, cellular uptake, and endocrine disruption at the maternal-fetal interface within the available literature.
Collapse
Affiliation(s)
- S Adams
- Department of Pharmacology and Toxicology, USA
| | - P A Stapleton
- Department of Pharmacology and Toxicology, USA; Environmental Occupational and Health Sciences Institute, Rutgers University, Piscataway, NJ, 08854, USA.
| |
Collapse
|