151
|
Placental Mitochondrial Function and Dysfunction in Preeclampsia. Int J Mol Sci 2023; 24:ijms24044177. [PMID: 36835587 PMCID: PMC9963167 DOI: 10.3390/ijms24044177] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The placenta is a vital organ of pregnancy, regulating adaptation to pregnancy, gestational parent/fetal exchange, and ultimately, fetal development and growth. Not surprisingly, in cases of placental dysfunction-where aspects of placental development or function become compromised-adverse pregnancy outcomes can result. One common placenta-mediated disorder of pregnancy is preeclampsia (PE), a hypertensive disorder of pregnancy with a highly heterogeneous clinical presentation. The wide array of clinical characteristics observed in pregnant individuals and neonates of a PE pregnancy are likely the result of distinct forms of placental pathology underlying the PE diagnosis, explaining why no one common intervention has proven effective in the prevention or treatment of PE. The historical paradigm of placental pathology in PE highlights an important role for utero-placental malperfusion, placental hypoxia and oxidative stress, and a critical role for placental mitochondrial dysfunction in the pathogenesis and progression of the disease. In the current review, the evidence of placental mitochondrial dysfunction in the context of PE will be summarized, highlighting how altered mitochondrial function may be a common feature across distinct PE subtypes. Further, advances in this field of study and therapeutic targeting of mitochondria as a promising intervention for PE will be discussed.
Collapse
|
152
|
Meakin C, Kim C, Lampert T, Aleksunes LM. High-throughput screening of toxicants that modulate extravillous trophoblast migration. Toxicol Lett 2023; 375:1-7. [PMID: 36535517 PMCID: PMC9877196 DOI: 10.1016/j.toxlet.2022.12.004] [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: 07/31/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Migration and subsequent invasion of extravillous trophoblasts into the uterus is essential for proper formation of the placenta. Disruption of these processes may result in poor pregnancy outcomes including preeclampsia, placenta accreta, fetal growth restriction, or fetal death. Currently, there are several methods for quantifying cell migration and invasion in vitro, each with limitations. Therefore, we developed a novel, high-throughput method to screen chemicals for their ability to alter human trophoblast migration. Human HTR8/SVneo trophoblast cells were cultured in Oris™ cell migration plates containing stopper barriers. After EVT cells attached and chemicals were added to media, stoppers were removed thereby creating a cell-free detection zone for migration. Entry of trophoblasts into this zone was monitored through imaging every 6 h and used to calculate a relative cell density. Chemicals known to increase (epidermal growth factor) and decrease (pertussis toxin and cadmium) trophoblast migration were used to validate this in vitro method. Next, a panel of environmental chemicals including bisphenols, mycoestrogens, and flame retardants, were screened for their ability to alter trophoblast invasion. In conclusion, a real-time method to track extravillous trophoblast migration offers potential for screening contaminants as placental toxicants.
Collapse
Affiliation(s)
- Cassandra Meakin
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Christine Kim
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | | | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA; Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
153
|
An SM, Kim MJ, Jeong JS, Kim SY, Kim DS, An BS, Kim SC. Oxytocin modulates steroidogenesis-associated genes and estradiol levels in the placenta. Syst Biol Reprod Med 2023; 69:223-233. [PMID: 36787388 DOI: 10.1080/19396368.2023.2170296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Oxytocin (OXT) plays a significant role during pregnancy, especially toward the end of pregnancy. Some studies have reported that OXT is involved in the stimulation of steroidogenesis in several organs. However, the effects of OXT on placental steroidogenesis have not yet been established. In this study, we investigated the regulation of steroid hormones and steroidogenic enzymes by OXT-associated signaling in vitro and in vivo. OXT increased the gene expression of steroidogenic enzymes, which convert pregnenolone to progesterone and dehydroepiandrosterone (DHEA) in vitro. In OXT-administered pregnant rats, pregnenolone and DHEA levels were significantly enhanced in the plasma and the expression of the enzymes synthesizing DHEA, testosterone, and estradiol (E2) was increased in placental tissues. Furthermore, OXT was found to affect placental cell differentiation, which is closely related to steroid hormone synthesis. After treatment of the pregnant rats with atosiban, an antagonist of the OXT receptor, the concentration of E2 in the plasma and the expression of E2-synthesizing enzyme were reduced. This regulation may be due to OXT-mediated differentiation, because OXT increases the expression of corticotropin-releasing hormone, which is a biomarker of placental cell differentiation. Our findings suggest that OXT contributes to maintaining pregnancy by regulating the differentiation of placental cells and steroidogenesis during pregnancy.
Collapse
Affiliation(s)
- Sung-Min An
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Gyeongsangnam-do, Republic of Korea
| | - Min Jae Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Gyeongsangnam-do, Republic of Korea
| | - Jea Sic Jeong
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Gyeongsangnam-do, Republic of Korea
| | - So Young Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Gyeongsangnam-do, Republic of Korea
| | - Da Som Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Gyeongsangnam-do, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science/Life and Industry Convergence Research Institute, Pusan National University, Miryang, Gyeongsangnam-do, Republic of Korea
| | - Seung Chul Kim
- Department of Obstetrics and Gynecology, Biomedical Research Institute, Pusan National University School of Medicine, Busan, Republic of Korea
| |
Collapse
|
154
|
Rudge MVC, Alves FCB, Hallur RLS, Oliveira RG, Vega S, Reyes DRA, Floriano JF, Prudencio CB, Garcia GA, Reis FVDS, Emanueli C, Fuentes G, Cornejo M, Toledo F, Valenzuela-Hinrichsen A, Guerra C, Grismaldo A, Valero P, Barbosa AMP, Sobrevia L. Consequences of the exposome to gestational diabetes mellitus. Biochim Biophys Acta Gen Subj 2023; 1867:130282. [PMID: 36436753 DOI: 10.1016/j.bbagen.2022.130282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/14/2022] [Accepted: 11/16/2022] [Indexed: 11/26/2022]
Abstract
The exposome is the cumulative measure of environmental influences and associated biological responses throughout the lifespan, including those from the environment, diet, behaviour, and endogenous processes. The exposome concept and the 2030 Agenda for the Sustainable Development Goals (SDGs) from the United Nations are the basis for understanding the aetiology and consequences of non-communicable diseases, including gestational diabetes mellitus (GDM). Pregnancy may be developed in an environment with adverse factors part of the immediate internal medium for fetus development and the external medium to which the pregnant woman is exposed. The placenta is the interface between maternal and fetal compartments and acts as a protective barrier or easing agent to transfer exposome from mother to fetus. Under and over-nutrition in utero, exposure to adverse environmental pollutants such as heavy metals, endocrine-disrupting chemicals, pesticides, drugs, pharmaceuticals, lifestyle, air pollutants, and tobacco smoke plays a determinant role in the development of GDM. This phenomenon is worsened by metabolic stress postnatally, such as obesity which increases the risk of GDM and other diseases. Clinical risk factors for GDM development include its aetiology. It is proposed that knowledge-based interventions to change the potential interdependent ecto-exposome and endo-exposome could avoid the occurrence and consequences of GDM.
Collapse
Affiliation(s)
- Marilza V C Rudge
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil.
| | - Fernanda C B Alves
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Raghavendra L S Hallur
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil; Centre for Biotechnology, Pravara Institute of Medical Sciences (DU), Loni-413736, Rahata Taluk, Ahmednagar District, Maharashtra, India
| | - Rafael G Oliveira
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Sofia Vega
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - David R A Reyes
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Juliana F Floriano
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Caroline B Prudencio
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Gabriela A Garcia
- São Paulo State University (UNESP), School of Sciences, Postgraduate Program in Materials Science and Technology (POSMAT), 17033-360 Bauru, São Paulo, Brazil
| | - Fabiana V D S Reis
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London SW3 6LY, UK
| | - Gonzalo Fuentes
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, The Netherlands; Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Marcelo Cornejo
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, The Netherlands; Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile; Faculty of Health Sciences, Universidad de Antofagasta, Antofagasta 02800, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Fernando Toledo
- Faculty of Basic Sciences, Universidad del Bío-Bío, Chillán 3780000, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Andrés Valenzuela-Hinrichsen
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Catalina Guerra
- Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Adriana Grismaldo
- Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León 64710, Mexico; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Paola Valero
- Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | - Angelica M P Barbosa
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil; Department of Physiotherapy and Occupational Therapy, School of Philosophy and Sciences, São Paulo State University (UNESP), 17525-900 Marília, São Paulo, Brazil
| | - Luis Sobrevia
- Department of Gynaecology and Obstetrics, Botucatu Medical School, São Paulo State University (UNESP), 18618-687 Botucatu, São Paulo, Brazil; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, The Netherlands; Tecnologico de Monterrey, Eutra, The Institute for Obesity Research (IOR), School of Medicine and Health Sciences, Monterrey, Nuevo León 64710, Mexico; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston QLD 4029, Queensland, Australia; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrician, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| |
Collapse
|
155
|
Adamova P, Lotto RR, Powell AK, Dykes IM. Are there foetal extracellular vesicles in maternal blood? Prospects for diagnostic biomarker discovery. J Mol Med (Berl) 2023; 101:65-81. [PMID: 36538060 PMCID: PMC9977902 DOI: 10.1007/s00109-022-02278-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 11/14/2022] [Accepted: 12/05/2022] [Indexed: 03/02/2023]
Abstract
Prenatal diagnosis of congenital disease improves clinical outcomes; however, as many as 50% of congenital heart disease cases are missed by current ultrasound screening methods. This indicates a need for improved screening technology. Extracellular vesicles (EVs) have attracted enormous interest in recent years for their potential in diagnostics. EVs mediate endocrine signalling in health and disease and are known to regulate aspects of embryonic development. Here, we critically evaluate recent evidence suggesting that EVs released from the foetus are able to cross the placenta and enter the maternal circulation. Furthermore, EVs from the mother appear to be transported in the reverse direction, whilst the placenta itself acts as a source of EVs. Experimental work utilising rodent models employing either transgenically encoded reporters or application of fluorescent tracking dyes provide convincing evidence of foetal-maternal crosstalk. This is supported by clinical data demonstrating expression of placental-origin EVs in maternal blood, as well as limited evidence for the presence of foetal-origin EVs. Together, this work raises the possibility that foetal EVs present in maternal blood could be used for the diagnosis of congenital disease. We discuss the challenges faced by researchers in translating these basic science findings into a clinical non-invasive prenatal test.
Collapse
Affiliation(s)
- Petra Adamova
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK.,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK
| | - Robyn R Lotto
- Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK.,School of Nursing and Allied Health, Liverpool John Moores University, Tithebarn St, Liverpool, L2 2ER, UK
| | - Andrew K Powell
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK.,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK
| | - Iain M Dykes
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom St, Liverpool, L3 3AF, UK. .,Liverpool Centre for Cardiovascular Science, Liverpool John Moores University, Liverpool, UK.
| |
Collapse
|
156
|
Mukherjee I, Singh S, Karmakar A, Kashyap N, Mridha AR, Sharma JB, Luthra K, Sharma RS, Biswas S, Dhar R, Karmakar S. New immune horizons in therapeutics and diagnostic approaches to Preeclampsia. Am J Reprod Immunol 2023; 89:e13670. [PMID: 36565013 DOI: 10.1111/aji.13670] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 11/02/2022] [Accepted: 12/10/2022] [Indexed: 12/25/2022] Open
Abstract
Hypertensive disorders of pregnancy (HDP) are one of the commonest maladies, affecting 5%-10% of pregnancies worldwide. The American College of Obstetricians and Gynecologists (ACOG) identifies four categories of HDP, namely gestational hypertension (GH), Preeclampsia (PE), chronic hypertension (CH), and CH with superimposed PE. PE is a multisystem, heterogeneous disorder that encompasses 2%-8% of all pregnancy-related complications, contributing to about 9% to 26% of maternal deaths in low-income countries and 16% in high-income countries. These translate to 50 000 maternal deaths and over 500 000 fetal deaths worldwide, therefore demanding high priority in understanding clinical presentation, screening, diagnostic criteria, and effective management. PE is accompanied by uteroplacental insufficiency leading to vascular and metabolic changes, vasoconstriction, and end-organ ischemia. PE is diagnosed after 20 weeks of pregnancy in women who were previously normotensive or hypertensive. Besides shallow trophoblast invasion and inadequate remodeling of uterine arteries, dysregulation of the nonimmune system has been the focal point in PE. This results from aberrant immune system activation and imbalanced differentiation of T cells. Further, a failure of tolerance toward the semi-allogenic fetus results due to altered distribution of Tregs such as CD4+FoxP3+ or CD4+CD25+CD127(low) FoxP3+ cells, thereby creating a cytotoxic environment by suboptimal production of immunosuppressive cytokines like IL-10, IL-4, and IL-13. Also, intracellular production of complement protein C5a may result in decreased FoxP3+ regulatory T cells. With immune system dysfunction as a major driver in PE pathogenesis, it is logical that therapeutic targeting of components of the immune system with pharmacologic agents like anti-inflammatory and immune-modulating molecules are either being used or under clinical trial. Cholesterol synthesis inhibitors like Pravastatin may improve placental perfusion in PE, while Eculizumab (monoclonal antibody inhibiting C5) and small molecular inhibitor of C5a, Zilucoplan are under investigation. Monoclonal antibody against IL-17(Secukinumab) has been proposed to alter the Th imbalance in PE. Autologous Treg therapy and immune checkpoint inhibitors like anti-CTLA-4 are emerging as new candidates in immune horizons for PE management in the future.
Collapse
Affiliation(s)
- Indrani Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.,Amity Institute of Biotechnology (AIB), Amity University, Noida, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Abhibrato Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Neha Kashyap
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Asit Ranjan Mridha
- Department of Obstetrics & Gynaecology, All India Institute of Medical Sciences, New Delhi, India
| | - Jai Bhagwan Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kalpana Luthra
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Radhey Shyam Sharma
- Ex-Head and Scientist G, Indian Council of Medical Research, New Delhi, India
| | - Subhrajit Biswas
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University, Noida, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
157
|
Sha L, Yong X, Shao Z, Duan Y, Hong Q, Zhang J, Zhang Y, Chen L. Targeting adverse effects of antiseizure medication on offspring: current evidence and new strategies for safety. Expert Rev Neurother 2023; 23:141-156. [PMID: 36731825 DOI: 10.1080/14737175.2023.2176751] [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: 02/04/2023]
Abstract
INTRODUCTION For women with epilepsy of reproductive age, antiseizure medications (ASMs) are associated with an increased risk of offspring malformations. There are safety concerns for most anti-seizure medications in the perinatal period, and there is a clear need to identify safe medications. ASMs must transport through biological barriers to exert toxic effects on the fetus, and transporters play essential roles in trans-barrier drug transport. Therefore, it is vital to understand the distribution and properties of ASM-related transporters in biological barriers. AREAS COVERED This study reviews the structure, transporter distribution, and properties of the blood-brain, placental, and blood-milk barrier, and summarizes the existing evidence for the trans-barrier transport mechanism of ASMs and standard experimental models of biological barriers. EXPERT OPINION Ideal ASMs in the perinatal period should have the following characteristics: 1) Increased transport through the blood-brain barrier, and 2) Reduced transport of the placental and blood-milk barriers. Thus, only low-dose or almost no antiseizure medication could enter the fetus's body, which could decrease medication-induced fetal abnormalities. Based on the stimulated structure and molecular docking, we propose a development strategy for new ASMs targeting transporters of biological barriers to improve the perinatal treatment of female patients with epilepsy.
Collapse
Affiliation(s)
- Leihao Sha
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| | - Xihao Yong
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yifei Duan
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| | - Qiulei Hong
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| | - Jifa Zhang
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| | - Yunwu Zhang
- The current form, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Lei Chen
- Department of Neurology, Joint Research Institution of Altitude Health, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan
| |
Collapse
|
158
|
The Effect of Short-Term Aspirin Administration during Programmed Frozen-Thawed Embryo Transfer on Pregnancy Outcomes and Complications. J Clin Med 2023; 12:jcm12031064. [PMID: 36769712 PMCID: PMC9918171 DOI: 10.3390/jcm12031064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/13/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Low-dose aspirin is widely used during frozen-embryo transfer (FET) cycles. Its anti-platelet property makes it a potentially useful drug for the prevention of hypertension disorders of pregnancy (HDP). However, the existing evidence about the effect of short-term aspirin administration on pregnancy outcomes is not clear. In our study, we retrospectively investigated women who had their first or second FET cycles at the Reproductive Hospital Affiliated with Shandong University from April 2017 to December 2020. A total of 4454 programmed FET cycles were recruited. According to whether aspirin was administrated in the protocols, the patients were divided into two groups: The Control group (n = 2793, 85 of them using donor sperm) and the Aspirin group (n = 1661, 35 of them using donor sperm). We analyzed the pregnancy outcomes and pregnancy complications of these cycles and observed similar live birth rates. We found that the short-term use of aspirin at a dosage of 50 mg per day for women undergoing programmed FET did not elevate the live birth rate or decrease the incidence of a series of pregnancy complications, including HDP. Based on our experience, short-term administration of low-dose aspirin may not improve the outcomes of young women undergoing frozen-thawed embryo transfer cycles.
Collapse
|
159
|
Lodefalk M, Chelslín F, Patriksson Karlsson J, Hansson SR. Placental Changes and Neuropsychological Development in Children-A Systematic Review. Cells 2023; 12:cells12030435. [PMID: 36766778 PMCID: PMC9913696 DOI: 10.3390/cells12030435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/31/2023] Open
Abstract
Placental dysfunction may increase the offspring's later-life disease risk. The objective of this systematic review was to describe associations between pathological placental changes and neuropsychological outcomes in children after the neonatal period. The inclusion criteria were human studies; original research; direct placental variables; neuropsychological outcomes; and analysis between their associations. The exclusion criterion was the offspring's age-0-28 days or >19 years. The MEDLINE and EMBASE databases were last searched in May 2022. We utilized the ROBINS-I for the risk of bias assessment and performed a narrative synthesis. In total, 3252 studies were identified, out of which 16 were included (i.e., a total of 15,862 participants). Half of the studies were performed on children with neonatal complications, and 75% of the studies reported an association between a placental change and an outcome; however, following the completion of the funnel plots, a risk of publication bias was indicated. The largest study described a small association between placental size and a risk of psychiatric symptoms in boys only. Inconsistency between the studies limited the evidence in this review. In general, no strong evidence was found for an association between pathological placental changes and childhood neuropsychological outcomes after the neonatal period. However, the association between placental size and mental health in boys indicates a placental sexual dimorphism, thereby suggesting an increased vulnerability for male fetuses.
Collapse
Affiliation(s)
- Maria Lodefalk
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
- University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
- Correspondence:
| | - Felix Chelslín
- Department of Pediatrics, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Johanna Patriksson Karlsson
- University Health Care Research Centre, Faculty of Medicine and Health, Örebro University, 701 82 Örebro, Sweden
| | - Stefan R. Hansson
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences Lund, Lund University, 221 00 Lund, Sweden
- Department of Obstetrics and Gynecology, Skåne University Hospital, 214 28 Malmö, Sweden
| |
Collapse
|
160
|
Chen Q, Shan D, Xie Y, Luo X, Wu Y, Chen Q, Dong R, Hu Y. Single cell RNA sequencing research in maternal fetal interface. Front Cell Dev Biol 2023; 10:1079961. [PMID: 36704195 PMCID: PMC9871254 DOI: 10.3389/fcell.2022.1079961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023] Open
Abstract
The maternal-fetal interface is an essential environment for embryonic growth and development, and a successful pregnancy depends on the dynamic balance of the microenvironment at the maternal-fetal interface. Single-cell sequencing, which unlike bulk sequencing that provides averaged data, is a robust method for interpreting the cellular and molecular landscape at single-cell resolution. With the support of single-cell sequencing, the issue of maternal-fetal interface heterogeneity during pregnancy has been more deeply elaborated and understood, which is important for a deeper understanding of physiological and pathological pregnancy. In this paper, we analyze the recent studies of single-cell transcriptomics in the maternal-fetal interface, and provide new directions for understanding and treating various pathological pregnancies.
Collapse
Affiliation(s)
- Qian Chen
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China,*Correspondence: Qian Chen, ; Yayi Hu,
| | - Dan Shan
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yupei Xie
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Xingrong Luo
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yuxia Wu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Qiuhe Chen
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ruihong Dong
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yayi Hu
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China,Qingbaijiang Maternal and Child Health Hospital, Chengdu, China,*Correspondence: Qian Chen, ; Yayi Hu,
| |
Collapse
|
161
|
Davenport BN, Jones HN, Wilson RL. Placental treatment with insulin-like growth factor 1 via nanoparticle differentially impacts vascular remodeling factors in guinea pig sub-placenta/decidua. Front Physiol 2023; 13:1055234. [PMID: 36685211 PMCID: PMC9845775 DOI: 10.3389/fphys.2022.1055234] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/13/2022] [Indexed: 01/06/2023] Open
Abstract
Clinically, fetal growth restriction (FGR) is only detectable in later gestation, despite pathophysiological establishment likely earlier in pregnancy. Additionally, there are no effective in utero treatment options for FGR. We have developed a nanoparticle to deliver human insulin-like 1 growth factor (hIGF-1) in a trophoblast-specific manner which results in increased expression of hIGF-1. IGF-1 signaling in the placenta regulates multiple developmental processes including trophoblast invasion and maternal vascular remodeling, both of which can be diminished in the FGR placenta. We aimed to determine the effects of short-term hIGF-1 nanoparticle treatment on sub-placenta/decidua trophoblast signaling mechanisms in FGR and under normal growth conditions. Using the guinea pig maternal nutrient restriction (MNR) model of FGR, ultrasound-guided, intra-placenta injections of hIGF-1 nanoparticle were performed at gestational day 30-33, and dams sacrificed 5 days later. Sub-placenta/decidua tissue was separated from placenta for further analyses. Western blot was used to analyze protein expression of ERK/AKT/mTOR signaling proteins (phospho-Erk (pERK), phospho-Akt (pAKT), raptor, rictor and deptor). qPCR was used to analyze gene expression of vascular/remodeling factors [vascular endothelial growth factor (Vegf), placenta growth factor (Pgf), platelet-derived growth factor (Pdgf)) and tight junction/adhesion proteins (claudin 5 (Cldn5), p-glycoprotein (Abcb1), occludin (Ocln) and tight junction protein 1 (Zo1)]. MNR reduced expression of pERK, PdgfB and Cldn5, and increased expression of Ocln and Zo1 in the sub-placenta/decidua. In MNR + hIGF1 nanoparticle sub-placenta/decidua, expression of PdgfB, Ocln and Zo1 was normalized, whilst pAkt, VegfB, Vegf receptor 1 and PdgfB receptor were increased compared to MNR. In contrast, hIGF-1 nanoparticle treatment of normal placentas reduced expression of pERK, raptor and increased expression of the mTOR inhibitor deptor. This was associated with reduced expression of VegfA, Plgf, and PdgfB. Here we have shown that the impact of hIGF-1 nanoparticle treatment is dependent on pregnancy environment. Under MNR/FGR, hIGF-1 nanoparticle treatment triggers increased expression of growth factors and normalization of EMT factors. However, under normal conditions, the response of the placenta is to decrease AKT/mTOR signaling and growth factor expression to achieve homeostasis.
Collapse
Affiliation(s)
- Baylea N. Davenport
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| | - Helen N. Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| | - Rebecca L. Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, United States
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
162
|
Manna S, Scheel J, Noone A, McElwain CJ, Scaife C, Gupta S, English J, McCarthy C, McCarthy FP. A proteomic profile of the healthy human placenta. Clin Proteomics 2023; 20:1. [PMID: 36593452 PMCID: PMC9808999 DOI: 10.1186/s12014-022-09388-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/09/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The placenta remains one of the least studied organs within the human body. Yet, placental dysfunction has been associated with various pregnancy complications leading to both maternal and fetal death and long-term health consequences. The aim of this study was to characterise the protein networks of healthy term placental sub-anatomical regions using label free quantification mass spectrometry. METHODS Three healthy placentae were sampled at five sample sites and each biopsy was dissected into maternal-, middle-, and fetal- sub-anatomical regions. Quadrupole-orbitrap mass spectrometer was used in data dependant analysis mode to identify 1859 unique proteins before detailed differential expression between regions. RESULTS Protein profiling identified 1081, 1086, and 1101 proteins in maternal, middle, and fetal sub-anatomical regions respectively. Differentially expressed proteins were identified considering the effect between sample site location and sub-anatomical region on protein expression. Of these, 374 differentially expressed proteins (Two-way ANOVA adjusted p-value < 0.05, HSD Tukey adjusted p-value 0.05) were identified between sample site locations and sub-anatomical regions. The placenta specific disease map NaviCenta ( https://www.sbi.uni-rostock.de/minerva/index.xhtml?id=NaviCenta ) was used to focus functional analysis results to the placenta specific context. Subsequently, functional analysis with a focus on senescence, and mitochondrial function were performed. Significant differences were observed between sub-anatomical regions in protein intensity and composition. A decrease in anti-senescent proteins within the maternal sub-anatomical region, and an increase in proteins associated with a switch from ATP to fatty acid consumption as a source of energy between middle and fetal sub-anatomical regions were observed. CONCLUSION These results suggest that normal proteomic variations exist within the anatomical structure of the placenta, thus recommending serial sectioning methodology for consistent placental research.
Collapse
Affiliation(s)
- Samprikta Manna
- grid.411916.a0000 0004 0617 6269Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, Cork, Ireland
| | - Julia Scheel
- grid.10493.3f0000000121858338Dept. of Systems Biology and Bioinformatics, University of Rostock, 18057 Rostock, Germany
| | - Aisling Noone
- grid.7872.a0000000123318773Department of Anatomy and Neuroscience, Western Gateway Building, University College Cork, Cork, Ireland
| | - Colm J. McElwain
- grid.7872.a0000000123318773Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| | - Caitriona Scaife
- grid.7886.10000 0001 0768 2743UCD Conway Institute for Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Shailendra Gupta
- grid.10493.3f0000000121858338Dept. of Systems Biology and Bioinformatics, University of Rostock, 18057 Rostock, Germany
| | - Jane English
- grid.512512.0INFANT Research Centre, Cork, Ireland ,grid.7872.a0000000123318773Department of Anatomy and Neuroscience, Western Gateway Building, University College Cork, Cork, Ireland
| | - Cathal McCarthy
- grid.7872.a0000000123318773Department of Pharmacology and Therapeutics, Western Gateway Building, University College Cork, Cork, Ireland
| | - Fergus P. McCarthy
- grid.411916.a0000 0004 0617 6269Department of Obstetrics and Gynaecology, Cork University Maternity Hospital, University College Cork, Cork, Ireland ,grid.512512.0INFANT Research Centre, Cork, Ireland
| |
Collapse
|
163
|
Cáceres D, Ochoa M, González-Ortiz M, Bravo K, Eugenín J. Effects of Prenatal Cannabinoids Exposure upon Placenta and Development of Respiratory Neural Circuits. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:199-232. [PMID: 37466775 DOI: 10.1007/978-3-031-32554-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Cannabis use has risen dangerously during pregnancy in the face of incipient therapeutic use and a growing perception of safety. The main psychoactive compound of the Cannabis sativa plant is the phytocannabinoid delta-9-tetrahydrocannabinol (A-9 THC), and its status as a teratogen is controversial. THC and its endogenous analogues, anandamide (AEA) and 2-AG, exert their actions through specific receptors (eCBr) that activate intracellular signaling pathways. CB1r and CB2r, also called classic cannabinoid receptors, together with their endogenous ligands and the enzymes that synthesize and degrade them, constitute the endocannabinoid system. This system is distributed ubiquitously in various central and peripheral tissues. Although the endocannabinoid system's most studied role is controlling the release of neurotransmitters in the central nervous system, the study of long-term exposure to cannabinoids on fetal development is not well known and is vital for understanding environmental or pathological embryo-fetal or postnatal conditions. Prenatal exposure to cannabinoids in animal models has induced changes in placental and embryo-fetal organs. Particularly, cannabinoids could influence both neural and nonneural tissues and induce embryo-fetal pathological conditions in critical processes such as neural respiratory control. This review aims at the acute and chronic effects of prenatal exposure to cannabinoids on placental function and the embryo-fetal neurodevelopment of the respiratory pattern. The information provided here will serve as a theoretical framework to critically evaluate the teratogen effects of the consumption of cannabis during pregnancy.
Collapse
Affiliation(s)
- Daniela Cáceres
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Martín Ochoa
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Marcelo González-Ortiz
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| | - Karina Bravo
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
- Facultad de Ingeniería, Universidad Autónoma de Chile, Providencia, Chile
| | - Jaime Eugenín
- Laboratorio de Sistemas Neurales, Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile.
| |
Collapse
|
164
|
Kruger L, Yue G, Paquette A, Sathyanarayana S, Enquobahrie DA, Bammler TK, MacDonald J, Zhao Q, Prasad B. An optimized proteomics-based approach to estimate blood contamination and cellular heterogeneity of frozen placental tissue. Placenta 2023; 131:111-118. [PMID: 36584637 PMCID: PMC9912121 DOI: 10.1016/j.placenta.2022.12.007] [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: 10/13/2022] [Revised: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Archived human placental tissue specimens are vital for studying placenta pathophysiology and toxicology. Proteomics analysis of placental tissue provides mechanistic and translational information, but the highly perfused and heterogenous nature of the placenta creates confounding technical variability. In this study, we developed an optimized proteomics-based approach to address the technical variability of proteomics data by normalizing blood contamination and cellular heterogeneity of archived placenta samples. METHODS Placenta samples (n = 99) were homogenized, digested using trypsin, and analyzed by liquid chromatography mass-spectrometry. Label-free quantification (LFQ) intensities of the proteins were analyzed for their correlation with blood (albumin) and placenta (aromatase) markers. Proteins that positively correlated with albumin and negatively correlated with aromatase or vice versa were considered blood and placental proteins, respectively. Next, the cellular heterogeneity of individual placenta samples was evaluated by quantifying specific cellular markers of cytotrophoblasts, syncytiotrophoblasts, extravillous trophoblasts, fibroblasts, Hofbauer cells, and decidual cells. RESULTS We found that placental proteins were contaminated by 41 to 85% blood proteins. Analysis of cellular markers confirmed syncytiotrophoblasts as the major cell type in placenta (i.e., 41 ± 9% of all cell types). Two samples showed distinct cell compositions with higher levels of the extravillous trophoblasts and decidual cells. DISCUSSION In summary, the optimized proteomics-based approach to estimate blood contamination and cellular heterogeneity of placental tissues has the potential to address technical variability in placenta proteomics analysis, which can be extended to other highly perfused and heterogenous tissues.
Collapse
Affiliation(s)
- Laken Kruger
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Guihua Yue
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Alison Paquette
- Seattle Children's Research Institute, Seattle, WA, USA; University of Washington, Seattle, WA, USA
| | - Sheela Sathyanarayana
- Seattle Children's Research Institute, Seattle, WA, USA; University of Washington, Seattle, WA, USA
| | | | | | | | - Qi Zhao
- University of Tennessee Health Science Center, Memphis, TN, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA.
| |
Collapse
|
165
|
Valverde-Pérez E, Prieto-Lloret J, Gonzalez-Obeso E, Cabero MI, Nieto ML, Pablos MI, Obeso A, Gomez-Niño A, Cárdaba-García RM, Rocher A, Olea E. Effects of Gestational Intermittent Hypoxia on Placental Morphology and Fetal Development in a Murine Model of Sleep Apnea. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1427:73-81. [PMID: 37322337 DOI: 10.1007/978-3-031-32371-3_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Obstructive sleep apnea (OSA) during pregnancy is characterized by episodes of intermittent hypoxia (IH) during sleep, resulting in adverse health outcomes for mother and offspring. Despite a prevalence of 8-20% in pregnant women, this disorder is often underdiagnosed.We have developed a murine model of gestational OSA to study IH effects on pregnant mothers, placentas, fetuses, and offspring. One group of pregnant rats was exposed to IH during the last 2 weeks of gestation (GIH). One day before the delivery date, a cesarean section was performed. Other group of pregnant rats was allowed to give birth at term to study offspring's evolution.Preliminary results showed no significant weight differences in mothers and fetuses. However, the weight of GIH male offspring was significantly lower than the controls at 14 days (p < 0.01). The morphological study of the placentas showed an increase in fetal capillary branching, expansion of maternal blood spaces, and number of cells of the external trophectoderm in the tissues from GIH-exposed mothers. Additionally, the placentas from the experimental males were enlarged (p < 0.05). Further studies are needed to follow the long-term evolution of these changes to relate the histological findings of the placentas with functional development of the offspring in adulthood.
Collapse
Affiliation(s)
- Esther Valverde-Pérez
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Jesús Prieto-Lloret
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Elvira Gonzalez-Obeso
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
- Servicio de Anatomía Patológica, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - María I Cabero
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Maria L Nieto
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Marta I Pablos
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
| | - Ana Obeso
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Angela Gomez-Niño
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
- Departamento de Biología Celular, Genética, Histología y Farmacología, Universidad de Valladolid, Valladolid, Spain
| | - Rosa M Cárdaba-García
- Departamento de Enfermería, Grupo de Investigación en Cuidados Enfermeros (GICE), Facultad de Enfermeria, Universidad de Valladolid, Valladolid, Spain
| | - Asunción Rocher
- Departamento de Bioquímica y Biología Molecular y Fisiología, Facultad de Medicina, Universidad de Valladolid, Valladolid, Spain
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain
| | - Elena Olea
- Instituto de Biomedicina y Genética Molecular (IBGM), UVa-CSIC, Valladolid, Spain.
- Departamento de Enfermería, Grupo de Investigación en Cuidados Enfermeros (GICE), Facultad de Enfermeria, Universidad de Valladolid, Valladolid, Spain.
| |
Collapse
|
166
|
Elkin ER, Su AL, Dou JF, Colacino JA, Bridges D, Padmanabhan V, Harris SM, Boldenow E, Loch-Caruso R, Bakulski KM. Sexually concordant and dimorphic transcriptional responses to maternal trichloroethylene and/or N-acetyl cysteine exposure in Wistar rat placental tissue. Toxicology 2023; 483:153371. [PMID: 36396003 PMCID: PMC10078828 DOI: 10.1016/j.tox.2022.153371] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/20/2022] [Accepted: 11/02/2022] [Indexed: 11/16/2022]
Abstract
Numerous Superfund sites are contaminated with the volatile organic chemical trichloroethylene (TCE). In women, exposure to TCE in pregnancy is associated with reduced birth weight. Our previous study reported that TCE exposure in pregnant rats decreased fetal weight and elevated oxidative stress biomarkers in placentae, suggesting placental injury as a potential mechanism of TCE-induced adverse birth outcomes. In this study, we investigated if co-exposure with the antioxidant N-acetylcysteine (NAC) attenuates TCE exposure effects on RNA expression. Timed-pregnant Wistar rats were exposed orally to 480 mg TCE/kg/day on gestation days 6-16. Exposure of 200 mg NAC/kg/day alone or as a pre/co-exposure with TCE occurred on gestation days 5-16 to stimulate antioxidant genes prior to TCE exposure. Tissue was collected on gestation day 16. In male and female placentae, we evaluated TCE- and/or NAC-induced changes to gene expression and pathway enrichment analyses using false discovery rate (FDR) and fold-change criteria. In female placentae, exposure to TCE caused significant differential expression 129 genes while the TCE+NAC altered 125 genes, compared with controls (FDR< 0.05 + fold-change >1). In contrast, in male placentae TCE exposure differentially expressed 9 genes and TCE+NAC differentially expressed 35 genes, compared with controls (FDR< 0.05 + fold-change >1). NAC alone did not significantly alter gene expression in either sex. Differentially expressed genes observed with TCE exposure were enriched in mitochondrial biogenesis and oxidative phosphorylation pathways in females whereas immune system pathways and endoplasmic reticulum stress pathways were differentially expressed in both sexes (FDR<0.05). TCE treatment was differentially enriched for genes regulated by the transcription factors ATF6 (both sexes) and ATF4 (males only), indicating a cellular condition triggered by misfolded proteins during endoplasmic reticulum stress. This study demonstrates novel genes and pathways involved in TCE-induced placental injury and showed antioxidant co-treatment largely did not attenuate TCE exposure effects.
Collapse
Affiliation(s)
- Elana R Elkin
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA.
| | - Anthony L Su
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - John F Dou
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Justin A Colacino
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA; Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA; Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA; Department of Pediatrics, Michigan Medicine, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, Michigan Medicine, Ann Arbor, MI, USA
| | - Sean M Harris
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Erica Boldenow
- Department of Biology, Calvin University, Grand Rapids, MI, USA
| | - Rita Loch-Caruso
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Kelly M Bakulski
- Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
167
|
Liang L, Chen Y, Wu C, Cao Z, Xia L, Meng J, He L, Yang C, Wang Z. MicroRNAs: key regulators of the trophoblast function in pregnancy disorders. J Assist Reprod Genet 2023; 40:3-17. [PMID: 36508034 PMCID: PMC9742672 DOI: 10.1007/s10815-022-02677-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
The placenta is essential for a successful pregnancy and healthy intrauterine development in mammals. During human pregnancy, the growth and development of the placenta are inseparable from the rapid proliferation, invasion, and migration of trophoblast cells. Previous reports have shown that the occurrence of many pregnancy disorders may be closely related to the dysfunction of trophoblasts. However, the function regulation of human trophoblast cells in the placenta is poorly understood. Therefore, studying the factors that regulate the function of trophoblast cells is necessary. MicroRNAs (miRNAs) are small, non-coding, single-stranded RNA molecules. Increasing evidence suggests that miRNAs play a crucial role in regulating trophoblast functions. This review outlines the role of miRNAs in regulating the function of trophoblast cells and several common signaling pathways related to miRNA regulation in pregnancy disorders.
Collapse
Affiliation(s)
- Lingli Liang
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| | - Yanjun Chen
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| | - Chunyan Wu
- grid.412017.10000 0001 0266 8918Department of Cardiovascular, The Third Affiliated Hospital of University of South China, Hengyang, 421001 China
| | - Zitong Cao
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| | - Linzhen Xia
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| | - Jun Meng
- grid.461579.8Department of Function, The First Affiliated Hospital of University of South China, Hengyang, 421001 China
| | - Lu He
- grid.461579.8Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang, 421001 China
| | - Chunfen Yang
- grid.461579.8Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang, 421001 China
| | - Zuo Wang
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| |
Collapse
|
168
|
Fresch R, Courtney J, Brockway H, Wilson RL, Jones H. HAND1 knockdown disrupts trophoblast global gene expression. Physiol Rep 2023; 11:e15553. [PMID: 36695714 PMCID: PMC9875743 DOI: 10.14814/phy2.15553] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 01/26/2023] Open
Abstract
Congenital heart disease (CHD) affects nearly 1% of births annually, and CHD pregnancies carry increased risk of developing pathologies of abnormal placentation. We previously reported significant developmental impacts of disrupting Hand1, a gene associated with CHD, expression in placenta trophoblast and endothelial cells in multiple mouse models. In this study, we aimed to build upon this knowledge and characterize the mechanistic impacts of disrupting HAND1 on human placenta trophoblast and vascular endothelial cell gene expression. HAND1 gene expression was silenced in BeWo cells, a choriocarcinoma model of human cytotrophoblasts, (n = 3-9 passages) and isolated human placental microvascular endothelial cells (HPMVEC; n = 3 passages), with HAND1 siRNA for 96 h. Cells were harvested, mRNA isolated and RNA sequencing performed using the Illumina NextSeq 550 platform. Normalization and differential gene expression analyses were conducted using general linear modeling in edgeR packages. Statistical significance was determined using a log2 fold change of >1.0 or < -1.0 and unadjusted p-value ≤0.05. Panther DB was used for overrepresentation analysis, and String DB for protein association network analysis. There was downregulation of 664 genes, and upregulation of 59 genes in BeWo cells with direct HAND1 knockdown. Overrepresentation analysis identified disruption to pathways including cell differentiation, localization, and cell projection organization. In contrast, only seven genes were changed with direct HAND1 knockdown in HPMVECs. Disruption to HAND1 expression significantly alters gene expression profile in trophoblast but not endothelial cells. This data provides further evidence that future studies on genetic perturbations in CHDs should consider the extra-embryonic tissue in addition to the fetal heart.
Collapse
Affiliation(s)
- Robert Fresch
- Division of Maternal‐Fetal Medicine, Department of Obstetrics and GynecologyUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Jennifer Courtney
- Center for Fetal and Placental ResearchCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Heather Brockway
- Center for Scientific ReviewNational Institutes of HealthBethesdaMarylandUSA
- Department of Physiology and AgingUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Rebecca L. Wilson
- Department of Physiology and AgingUniversity of Florida College of MedicineGainesvilleFloridaUSA
- Center for Research in Perinatal OutcomesUniversity of Florida College of MedicineGainesvilleFloridaUSA
| | - Helen Jones
- Department of Physiology and AgingUniversity of Florida College of MedicineGainesvilleFloridaUSA
- Center for Research in Perinatal OutcomesUniversity of Florida College of MedicineGainesvilleFloridaUSA
| |
Collapse
|
169
|
Kravchenko OV, Tiulienieva OA, Yasnikovska SM, Goshovska AV. FEATURES OF THE FUNCTIONAL MORPHOLOGY OF THE FULL-TERM PLACENTA IN WOMEN WITH THREATENED ABORTION WITH BLEEDING IN THE FIRST TRIMESTER OF GESTATION. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2023; 76:1797-1803. [PMID: 37740973 DOI: 10.36740/wlek202308114] [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: 09/25/2023]
Abstract
OBJECTIVE The aim: To examine the morphology regarding the term placentas in pregnancies with threatened abortion with bleeding in the first trimester. PATIENTS AND METHODS Materials and methods: 118 term placentas were selected, of which 40 placentas with the physiological course of pregnancy. 78 placentas were from women with threatened abortion with bleeding in the first trimester, of which 37 patients received hormonal therapy (group I), 41 women were prescribed symptomatic therapy (group II). Placentas were investigatedaccording to the protocol, which includes organometric, macroscopic and microscopic studies. RESULTS Results: In the placentas of group II is a significant increase of the area of terminal villi compared due to the stroma against the background of a deficit of fetal capillaries. In group I have revealed that the specific weight of the vascular bed of the terminal villi was 1.5 times higher compared to the control (Р=0.031) and 2.7 times higher than the group II (Р=0.022) and dominates the share of the stroma. The weight of the epithelium of the terminal villi in all groups is approximately the same (Р=0.042), but the ratio of the epithelium to the stroma is higher in the group I (0.63) than in the control (0.43). CONCLUSION Conclusions: In women with a pathological course of the first trimester of pregnancy the compensatory mechanisms in full-term placentas are morphologically represented by an increase in the number of terminal villi, syncytio-capillary membranes, intensification of angiogenesis. In the placentas of women who received hormonal therapy adaptive reactions are most effective and able to compensate for the gestational immaturity of the chorion.
Collapse
|
170
|
Inkster AM, Konwar C, Peñaherrera MS, Brain U, Khan A, Price EM, Schuetz JM, Portales-Casamar É, Burt A, Marsit CJ, Vaillancourt C, Oberlander TF, Robinson WP. Profiling placental DNA methylation associated with maternal SSRI treatment during pregnancy. Sci Rep 2022; 12:22576. [PMID: 36585414 PMCID: PMC9803674 DOI: 10.1038/s41598-022-26071-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022] Open
Abstract
Selective serotonin reuptake inhibitors (SSRIs) for treatment of prenatal maternal depression have been associated with neonatal neurobehavioral disturbances, though the molecular mechanisms remain poorly understood. In utero exposure to SSRIs may affect DNA methylation (DNAme) in the human placenta, an epigenetic mark that is established during development and is associated with gene expression. Chorionic villus samples from 64 human placentas were profiled with the Illumina MethylationEPIC BeadChip; clinical assessments of maternal mood and SSRI treatment records were collected at multiple time points during pregnancy. Case distribution was 20 SSRI-exposed cases and 44 SSRI non-exposed cases. Maternal depression was defined using a mean maternal Hamilton Depression score > 8 to indicate symptomatic depressed mood ("maternally-depressed"), and we further classified cases into SSRI-exposed, maternally-depressed (n = 14); SSRI-exposed, not maternally-depressed (n = 6); SSRI non-exposed, maternally-depressed (n = 20); and SSRI non-exposed, not maternally-depressed (n = 24). For replication, Illumina 450K DNAme profiles were obtained from 34 additional cases from an independent cohort (n = 17 SSRI-exposed, n = 17 SSRI non-exposed). No CpGs were differentially methylated at FDR < 0.05 comparing SSRI-exposed to non-exposed placentas, in a model adjusted for mean maternal Hamilton Depression score, or in a model restricted to maternally-depressed cases with and without SSRI exposure. However, at a relaxed threshold of FDR < 0.25, five CpGs were differentially methylated (|Δβ| > 0.03) by SSRI exposure status. Four were covered by the replication cohort measured by the 450K array, but none replicated. No CpGs were differentially methylated (FDR < 0.25) comparing maternally depressed to not depressed cases. In sex-stratified analyses for SSRI-exposed versus non-exposed cases (females n = 31; males n = 33), three additional CpGs in females, but none in males, were differentially methylated at the relaxed FDR < 0.25 cut-off. We did not observe large-scale alterations of DNAme in placentas exposed to maternal SSRI treatment, as compared to placentas with no SSRI exposure. We also found no evidence for altered DNAme in maternal depression-exposed versus depression non-exposed placentas. This novel work in a prospectively-recruited cohort with clinician-ascertained SSRI exposure and mood assessments would benefit from future replication.
Collapse
Affiliation(s)
- Amy M. Inkster
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Chaini Konwar
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics, Vancouver, BC V6H 0B3 Canada
| | - Maria S. Peñaherrera
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Ursula Brain
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada
| | - Almas Khan
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pediatrics, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - E. Magda Price
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3 Canada ,grid.28046.380000 0001 2182 2255Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON K1H 5B2 Canada
| | - Johanna M. Schuetz
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Élodie Portales-Casamar
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830Department of Pediatrics, University of British Columbia, Vancouver, BC V6T 1Z4 Canada
| | - Amber Burt
- grid.189967.80000 0001 0941 6502Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322 USA
| | - Carmen J. Marsit
- grid.189967.80000 0001 0941 6502Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322 USA
| | - Cathy Vaillancourt
- grid.418084.10000 0000 9582 2314INRS-Centre Armand Frappier and Réseau intersectoriel de recherche en santé de l’Université du Québec, Laval, QC H7V 1B7 Canada
| | - Tim F. Oberlander
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Wendy P. Robinson
- grid.414137.40000 0001 0684 7788BC Children’s Hospital Research Institute (BCCHR), 950 W 28th Ave, Vancouver, BC V5Z 4H4 Canada ,grid.17091.3e0000 0001 2288 9830Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| |
Collapse
|
171
|
Maternal Dexamethasone Exposure Induces Sex-Specific Changes in Histomorphology and Redox Homeostasis of Rat Placenta. Int J Mol Sci 2022; 24:ijms24010540. [PMID: 36613982 PMCID: PMC9820254 DOI: 10.3390/ijms24010540] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/30/2022] Open
Abstract
As the mediator between the mother and fetus, the placenta allows the most appropriate environment and optimal fetal growth. The placenta of one sex sometimes has a greater ability over the other to respond to and protect against possible maternal insults. Here, we characterized sex differences in the placenta’s morphological features and antioxidant status following dexamethasone (Dx) exposure. Pregnant rats were exposed to Dx or saline. The placenta was histologically and stereologically analyzed. The activity of the antioxidant enzymes, lipid peroxides (TBARS), superoxide anion and nitric oxide (NO) was measured. The decrease in placental zone volumes was more pronounced (p < 0.05) in female placentas. The volume density of PCNA-immunopositive nuclei was reduced (p < 0.05) in both sexes. The reduced (p < 0.05) antioxidant enzyme activities, enhanced TBARS and NO concentration indicate that Dx exposure triggered oxidative stress in the placenta of both fetal sexes, albeit stronger in the placenta of female fetuses. In conclusion, maternal Dx treatment reduced the size and volume of placental zones, altered placental histomorphology, decreased cell proliferation and triggered oxidative stress; however, the placentas of female fetuses exerted more significant responses to the treatment effects. The reduced placental size most probably reduced the transport of nutrients and oxygen, thus resulting in the reduced weight of fetuses, similar in both sexes. The lesser ability of the male placenta to detect and react to maternal exposure to environmental challenges may lead to long-standing health effects.
Collapse
|
172
|
Chen X, Tang AT, Tober J, Yang J, Leu NA, Sterling S, Chen M, Yang Y, Mericko-Ishizuka P, Speck NA, Kahn ML. Mouse placenta fetal macrophages arise from endothelial cells outside the placenta. Dev Cell 2022; 57:2652-2660.e3. [PMID: 36473461 PMCID: PMC9752200 DOI: 10.1016/j.devcel.2022.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/07/2022] [Accepted: 11/03/2022] [Indexed: 12/12/2022]
Abstract
Placental fetal macrophages (fMacs) are the only immune cells on the fetal side of the placental barrier. Mouse models have not been used to test their function because they have previously been found to have distinct cellular origins and functions in mice and humans. Here, we test the ontogeny of mouse placental fMacs. Using a new Hoxa13Cre allele that labels all placental endothelial cells (ECs), we demonstrate that mouse placenta fMacs do not arise from placental endothelium. Instead, lineage tracing studies using Tie2-Cre and Cx3cr1CreERT2 alleles demonstrate that mouse placental fMacs arise from yolk sac endothelium. Administration of blocking antibodies against CSF1R at E6.5 and E7.5 results in depletion of placental fMacs throughout pregnancy, and this suggests a yolk sac origin, similar to that in human fMacs. This Matters Arising paper is in response to Liang et al., published in Developmental Cell. A response by Liang and Liu is published in this issue.
Collapse
Affiliation(s)
- Xiaowen Chen
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alan T Tang
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joanna Tober
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jisheng Yang
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - N Adrian Leu
- Transgenic Mouse Core, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephanie Sterling
- Transgenic Mouse Core, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mei Chen
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yiqing Yang
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patricia Mericko-Ishizuka
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nancy A Speck
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark L Kahn
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
173
|
Perić M, Bečeheli I, Čičin-Šain L, Desoye G, Štefulj J. Serotonin system in the human placenta - the knowns and unknowns. Front Endocrinol (Lausanne) 2022; 13:1061317. [PMID: 36531448 PMCID: PMC9751904 DOI: 10.3389/fendo.2022.1061317] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
The biogenic monoamine serotonin (5-hydroxytryptamine, 5-HT) is a chemical messenger widely distributed in the brain and various other organs. Its homeostasis is maintained by the coordinated activity of a variety of proteins, including enzymes of serotonin metabolism, transmembrane transporters of serotonin, and serotonin receptors. The serotonin system has been identified also in the placenta in rodent models as a key component of placental physiology. However, serotonin pathways in the human placenta are far from well understood. Their alterations may have long-lasting consequences for the fetus that can manifest later in life. In this review, we summarize information on the location of the components of the serotonin system in the human placenta, their regulation, function, and alterations in pathological pregnancies. We highlight current controversies and discuss important topics for future research.
Collapse
Affiliation(s)
- Maja Perić
- Laboratory of Neurochemistry and Molecular Neurobiology, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Ivona Bečeheli
- Laboratory of Neurochemistry and Molecular Neurobiology, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Lipa Čičin-Šain
- Laboratory of Neurochemistry and Molecular Neurobiology, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Graz, Austria
| | - Jasminka Štefulj
- Laboratory of Neurochemistry and Molecular Neurobiology, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
174
|
Hollar DW. The competition of ecological resonances in the quantum metabolic model of cancer: Potential energetic interventions. Biosystems 2022; 222:104798. [DOI: 10.1016/j.biosystems.2022.104798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 11/02/2022]
|
175
|
Luconi M, Sogorb MA, Markert UR, Benfenati E, May T, Wolbank S, Roncaglioni A, Schmidt A, Straccia M, Tait S. Human-Based New Approach Methodologies in Developmental Toxicity Testing: A Step Ahead from the State of the Art with a Feto-Placental Organ-on-Chip Platform. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:15828. [PMID: 36497907 PMCID: PMC9737555 DOI: 10.3390/ijerph192315828] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/20/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Developmental toxicity testing urgently requires the implementation of human-relevant new approach methodologies (NAMs) that better recapitulate the peculiar nature of human physiology during pregnancy, especially the placenta and the maternal/fetal interface, which represent a key stage for human lifelong health. Fit-for-purpose NAMs for the placental-fetal interface are desirable to improve the biological knowledge of environmental exposure at the molecular level and to reduce the high cost, time and ethical impact of animal studies. This article reviews the state of the art on the available in vitro (placental, fetal and amniotic cell-based systems) and in silico NAMs of human relevance for developmental toxicity testing purposes; in addition, we considered available Adverse Outcome Pathways related to developmental toxicity. The OECD TG 414 for the identification and assessment of deleterious effects of prenatal exposure to chemicals on developing organisms will be discussed to delineate the regulatory context and to better debate what is missing and needed in the context of the Developmental Origins of Health and Disease hypothesis to significantly improve this sector. Starting from this analysis, the development of a novel human feto-placental organ-on-chip platform will be introduced as an innovative future alternative tool for developmental toxicity testing, considering possible implementation and validation strategies to overcome the limitation of the current animal studies and NAMs available in regulatory toxicology and in the biomedical field.
Collapse
Affiliation(s)
- Michaela Luconi
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy
- I.N.B.B. (Istituto Nazionale Biostrutture e Biosistemi), Viale Medaglie d’Oro 305, 00136 Rome, Italy
| | - Miguel A. Sogorb
- Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Avenida de la Universidad s/n, 03202 Elche, Spain
| | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Emilio Benfenati
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Tobias May
- InSCREENeX GmbH, Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Susanne Wolbank
- Ludwig Boltzmann Institut for Traumatology, The Research Center in Cooperation with AUVA, Austrian Cluster for Tissue Regeneration, Donaueschingenstrasse 13, 1200 Vienna, Austria
| | - Alessandra Roncaglioni
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milan, Italy
| | - Astrid Schmidt
- Placenta Lab, Department of Obstetrics, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Marco Straccia
- FRESCI by Science&Strategy SL, C/Roure Monjo 33, Vacarisses, 08233 Barcelona, Spain
| | - Sabrina Tait
- Centre for Gender-Specific Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| |
Collapse
|
176
|
Ortega MA, Fraile-Martínez O, García-Montero C, Paradela A, Asunción Sánchez-Gil M, Rodriguez-Martin S, De León-Luis JA, Pereda-Cerquella C, Bujan J, Guijarro LG, Alvarez-Mon M, García-Honduvilla N. Unfolding the role of placental-derived Extracellular Vesicles in Pregnancy: From homeostasis to pathophysiology. Front Cell Dev Biol 2022; 10:1060850. [PMID: 36478738 PMCID: PMC9720121 DOI: 10.3389/fcell.2022.1060850] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/02/2022] [Indexed: 08/11/2023] Open
Abstract
The human placenta is a critical structure with multiple roles in pregnancy, including fetal nutrition and support, immunological, mechanical and chemical barrier as well as an endocrine activity. Besides, a growing body of evidence highlight the relevance of this organ on the maternofetal wellbeing not only during gestation, but also from birth onwards. Extracellular vesicles (EVs) are complex macromolecular structures of different size and content, acting as carriers of a diverse set of molecules and information from donor to recipient cells. Since its early development, the production and function of placental-derived EVs are essential to ensure an adequate progress of pregnancy. In turn, the fetus receives and produce their own EVs, highlighting the importance of these components in the maternofetal communication. Moreover, several studies have shown the clinical relevance of EVs in different obstetric pathologies such as preeclampsia, infectious diseases or gestational diabetes, among others, suggesting that they could be used as pathophysiological biomarkers of these diseases. Overall, the aim of this article is to present an updated review of the published basic and translational knowledge focusing on the role of placental-derived EVs in normal and pathological pregnancies. We suggest as well future lines of research to take in this novel and promising field.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, Alcala de Henares, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | | | - María Asunción Sánchez-Gil
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- University Defense Center of Madrid (CUD), Madrid, Spain
| | - Sonia Rodriguez-Martin
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Service of Pediatric, Hospital Universitario Principe de Asturias, Alcalá de Henares, Spain
| | - Juan A. De León-Luis
- Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Ma-drid, Madrid, Spain
| | - Claude Pereda-Cerquella
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Luis G. Guijarro
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Unit of Biochemistry and Molecular Biology, Centro de Investigación Biomédica en Red en El Área Temática de Enfermedades Hepáticas (CIBEREHD), Department of System Biology, University of Alcalá, Alcala de Henares, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, Centro de Investigación Biomédica en Red en El Área Temática de Enfermedades Hepáticas (CIBEREHD), University Hospital Príncipe de Asturias, Alcala de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- University Defense Center of Madrid (CUD), Madrid, Spain
| |
Collapse
|
177
|
Ruan D, Ye ZW, Yuan S, Li Z, Zhang W, Ong CP, Tang K, Ka Ki Tam TT, Guo J, Xuan Y, Huang Y, Zhang Q, Lee CL, Lu L, Chiu PCN, Yeung WSB, Liu F, Jin DY, Liu P. Human early syncytiotrophoblasts are highly susceptible to SARS-CoV-2 infection. Cell Rep Med 2022; 3:100849. [PMID: 36495872 PMCID: PMC9671691 DOI: 10.1016/j.xcrm.2022.100849] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 08/23/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
Direct in vivo investigation of human placenta trophoblast's susceptibility to SARS-CoV-2 is challenging. Here we report that human trophoblast stem cells (hTSCs) and their derivatives are susceptible to SARS-CoV-2 infection, which reveals heterogeneity in hTSC cultures. Early syncytiotrophoblasts (eSTBs) generated from hTSCs have enriched transcriptomic features of peri-implantation trophoblasts, express high levels of angiotensin-converting enzyme 2 (ACE2), and are productively infected by SARS-CoV-2 and its Delta and Omicron variants to produce virions. Antiviral drugs suppress SARS-CoV-2 replication in eSTBs and antagonize the virus-induced blockage of STB maturation. Although less susceptible to SARS-CoV-2 infection, trophoblast organoids originating from hTSCs show detectable viral replication reminiscent of the uncommon placental infection. These findings implicate possible risk of COVID-19 infection in peri-implantation embryos, which may go unnoticed. Stem cell-derived human trophoblasts such as eSTBs can potentially provide unlimited amounts of normal and genome-edited cells and facilitate coronavirus research and antiviral discovery.
Collapse
Affiliation(s)
- Degong Ruan
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zi-Wei Ye
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Shuofeng Yuan
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Zhuoxuan Li
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Weiyu Zhang
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Chon Phin Ong
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kaiming Tang
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Timothy Theodore Ka Ki Tam
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jilong Guo
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yiyi Xuan
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yunying Huang
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Qingqing Zhang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Cheuk-Lun Lee
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Liming Lu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Philip C N Chiu
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - William S B Yeung
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Department of Obstetrics and Gynaecology, School of Clinical Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Fang Liu
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Foshan Stomatology Hospital, School of Medicine, Foshan University, No. 5 Hebing Road, Foshan, Guangdong Province, China.
| | - Dong-Yan Jin
- Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Pentao Liu
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China; Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
178
|
Schreiner C, Powell TL, Palmer C, Jansson T. Placental proteins with predicted roles in fetal development decrease in premature infants. Pediatr Res 2022; 92:1316-1324. [PMID: 35132128 PMCID: PMC9357234 DOI: 10.1038/s41390-022-01942-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/15/2021] [Accepted: 12/23/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Emerging evidence from animal experiments indicate that factors secreted by the placenta are critical for normal fetal organ development. Our objective was to characterize the umbilical vein and artery proteome in preterm infants and identify proteins that decrease in the neonatal circulation following delivery. METHODS Cord blood at delivery and neonatal blood at 48-72 h of life was collected in 25 preterm infants. Plasma protein abundance was determined using the SomaLogic platform. RESULTS When comparing protein levels of umbilical venous to arterial cord blood, 434 proteins were significantly higher indicating placental secretion into the fetal circulation. Moreover, when comparing neonatal blood to umbilical vein levels, 142 proteins were significantly lower. These proteins included Endoplasmic reticulum resident protein 29, CD59, Fibroblast growth factor 2 and Dynactin subunit 2, which are involved in brain development and prevention of brain damage as well as Fibroblast growth factor 1 which prevents lung fibrosis. CONCLUSIONS The late second trimester human placenta secretes proteins into the fetal circulation which decrease following delivery. Many of these proteins are predicted to be important in the development of fetal organs. Further studies are needed to directly link placental proteins to organ development and poor outcomes in preterm infants. IMPACT Prematurity remains a leading cause of morbidity and mortality requiring the development of novel treatments. Emerging evidence from animal studies suggest that factors secreted from the placenta may be critical in the development of the fetus. We report that the preterm human placenta secretes an array of proteins into the fetal circulation. Some of these proteins are predicted to be involved in the development of the brain and the lung. When born prematurely, infants are deprived of these placental proteins, which may contribute to their poor outcomes.
Collapse
Affiliation(s)
- Cynthia Schreiner
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Department of Pediatrics at Renown Children's Hospital, Reno, NV, USA.
| | - Theresa L Powell
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Department of Obstetrics, Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Claire Palmer
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Thomas Jansson
- Department of Obstetrics, Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
179
|
Espinoza ML, Brundler MA, Hasan SU, Mohammad K, Momin S, Al Shaikh B, Yusuf K. Placental pathology as a marker of brain injury in infants with hypoxic ischemic encephalopathy. Early Hum Dev 2022; 174:105683. [PMID: 36215816 DOI: 10.1016/j.earlhumdev.2022.105683] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/27/2022] [Accepted: 09/28/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Hypoxic Ischemic Encephalopathy (HIE) can lead to devastating consequences for the affected infant. Although therapeutic cooling benefits infants with moderate and severe HIE, differentiating mild from moderate-severe HIE may be challenging. The placenta reflects the fetal intrauterine environment and may reveal underlying processes that affect brain injury. AIM To describe placental histopathology using the Amsterdam Placental Workshop Group Criteria in different grades of HIE. STUDY DESIGN Retrospective cohort. SUBJECTS Infants admitted to a tertiary care neonatal intensive care unit with a diagnosis of HIE between 2011 and 2016. OUTCOME MEASURE Maternal and neonatal clinical variables and placental histopathology using the Amsterdam Placental Workshop Group Criteria were compared between mild and moderate-severe HIE. Mann-Whitney or t-test or ꭓ2 were performed for bivariate associations as appropriate. To explain the relationship between placental pathology and severity of HIE odds ratios (ORs) and 95 % confidence intervals (CIs) were calculated using logistic regression models. RESULTS Of the 73 infants in the study, 23 had mild and 50 moderate-sever HIE. There was no difference in maternal and neonatal characteristics except for sentinel events which were higher in the moderate- severe group. On placental histopathology, acute inflammation, including fetal inflammatory reaction (FIR) were significantly higher in the moderate-severe group. After adjusting for confounders, FIR remained significantly associated with moderate-severe HIE, ORs 6.29, 95 % CI 1.5-25. CONCLUSION Our study demonstrates FIR in the placenta is associated with severity of HIE.
Collapse
Affiliation(s)
- Maria Liza Espinoza
- Department of Pediatrics, University of Calgary, Section of Neonatology, Calgary, AB, Canada
| | - Marie-Anne Brundler
- Department of Pathology & Laboratory Medicine and Pediatrics, University of Calgary, Calgary, AB, Canada
| | - Shabih U Hasan
- Department of Pediatrics, University of Calgary, Section of Neonatology, Calgary, AB, Canada
| | - Khorshid Mohammad
- Department of Pediatrics, University of Calgary, Section of Neonatology, Calgary, AB, Canada
| | - Sarfaraz Momin
- Department of Pediatrics, University of Calgary, Section of Neonatology, Calgary, AB, Canada
| | - Belal Al Shaikh
- Department of Pediatrics, University of Calgary, Section of Neonatology, Calgary, AB, Canada
| | - Kamran Yusuf
- Department of Pediatrics, University of Calgary, Section of Neonatology, Calgary, AB, Canada.
| |
Collapse
|
180
|
Mekler T, Plitman Mayo R, Weissmann J, Marom G. Impact of tissue porosity and asymmetry on the oxygen uptake of the human placenta: A numerical study. Placenta 2022; 129:15-22. [PMID: 36183458 DOI: 10.1016/j.placenta.2022.09.008] [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: 04/28/2022] [Revised: 08/17/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION This study proposes a computational fluid dynamics model of a human placenta's independent exchange unit (placentome) to assess the effect that the inner villi distribution and decidual veins (DVs) location and number, have on the oxygen uptake. METHODS The internal placentome porosity distribution was altered in symmetric morphology, while asymmetry was introduced by varying the location and number of DVs. The DV asymmetry was introduced by either displacing them circumferentially, thereby changing the angle between them, or by adding DVs in the inlet cross-section. The results were analyzed by the changes in the normalized oxygen mass fraction and the oxygen uptake. RESULTS Oxygenated blood was shown to be delivered deeper into the placentome when the area of non-homogeneous porosity was larger. The largest oxygen uptake was achieved in the asymmetric model with the smallest angle distance between the DVs, where a 10% decrease relative to the farthest case was obtained. Placing DVs adjacent to the spiral artery opening enhanced the drainage of oxygenated blood. DISCUSSION This study demonstrates the importance of the local porosity distribution for the proper perfusion of the intervillous space and proposes a novel approach to improve our understanding of the role of the DVs in placental oxygen uptake.
Collapse
Affiliation(s)
- Tirosh Mekler
- School of Mechanical Engineering, Tel Aviv University, Tel Aviv, 6997801, Israel.
| | - Romina Plitman Mayo
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| | - Jonathan Weissmann
- Department of Biomedical Engineering, The Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 6997801, Israel.
| | - Gil Marom
- School of Mechanical Engineering, Tel Aviv University, Tel Aviv, 6997801, Israel.
| |
Collapse
|
181
|
Wilson RL, Lampe K, Gupta MK, Duvall CL, Jones HN. Nanoparticle-mediated transgene expression of insulin-like growth factor 1 in the growth restricted guinea pig placenta increases placenta nutrient transporter expression and fetal glucose concentrations. Mol Reprod Dev 2022; 89:540-553. [PMID: 36094907 PMCID: PMC10947605 DOI: 10.1002/mrd.23644] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 12/25/2022]
Abstract
Fetal growth restriction (FGR) significantly contributes to neonatal and perinatal morbidity and mortality. Currently, there are no effective treatment options for FGR during pregnancy. We have developed a nanoparticle gene therapy targeting the placenta to increase expression of human insulin-like growth factor 1 (hIGF1) to correct fetal growth trajectories. Using the maternal nutrient restriction guinea pig model of FGR, an ultrasound-guided, intraplacental injection of nonviral, polymer-based hIGF1 nanoparticle containing plasmid with the hIGF1 gene and placenta-specific Cyp19a1 promotor was administered at mid-pregnancy. Sustained hIGF1 expression was confirmed in the placenta 5 days after treatment. Whilst increased hIGF1 did not change fetal weight, circulating fetal glucose concentration were 33%-67% higher. This was associated with increased expression of glucose and amino acid transporters in the placenta. Additionally, hIGF1 nanoparticle treatment increased the fetal capillary volume density in the placenta, and reduced interhaemal distance between maternal and fetal circulation. Overall, our findings, that trophoblast-specific increased expression of hIGF1 results in changes to glucose transporter expression and increases fetal glucose concentrations within a short time period, highlights the translational potential this treatment could have in correcting impaired placental nutrient transport in human pregnancies complicated by FGR.
Collapse
Affiliation(s)
- Rebecca L. Wilson
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Kristin Lampe
- Center for Fetal and Placental Research, Cincinnati Children’s Hospital and Medical Center, Cincinnati, Ohio, USA
| | - Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Craig L. Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Helen N. Jones
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, Florida, USA
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, Gainesville, Florida, USA
| |
Collapse
|
182
|
Zheng Y, Pan J, Xia C, Chen H, Zhou H, Ju W, Wegiel J, Myatt L, Roberts JM, Guo X, Zhong N. Characterization of placental and decidual cell development in early pregnancy loss by single-cell RNA sequencing. Cell Biosci 2022; 12:168. [PMID: 36209198 PMCID: PMC9548121 DOI: 10.1186/s13578-022-00904-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 09/24/2022] [Indexed: 11/06/2022] Open
Abstract
Background Early pregnancy loss (EPL) presents as sporadic or recurrent miscarriage during the first trimester. In addition to chromosomal defects, EPL may result from impairment of the placental-decidual interface at early gestational age due to gene-environmental interactions. Methods To better understand the pathogenesis associated with this impairment, cell development in chorionic villi and decidua of different forms of EPL (sporadic or recurrent) was investigated with single-cell RNA sequencing and compared to that of normal first-trimester tissue. Results Unique gene expression signatures were obtained for the different forms of EPL and for normal tissue and the composition of placental and decidual cell clusters in each form was established. In particular, the involvement of macrophages in the EPL phenotypes was identified revealing an immunoactive state. Conclusion Differential gene expression and unique marker genes among cell clusters from chorionic villi and decidua of miscarried and normal pregnancies, may lead to identification of biomarker for EPL. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-022-00904-5.
Collapse
Affiliation(s)
- Yuhua Zheng
- Maternity and Child Healthcare Hospital, Foshan Women and Children, 11 W. Renmin Lu, Foshan, 528000 China
| | - Jing Pan
- Maternity and Child Healthcare Hospital, Foshan Women and Children, 11 W. Renmin Lu, Foshan, 528000 China
| | - Chenglai Xia
- Maternity and Child Healthcare Hospital, Foshan Women and Children, 11 W. Renmin Lu, Foshan, 528000 China
| | - Haiying Chen
- Maternity and Child Healthcare Hospital, Foshan Women and Children, 11 W. Renmin Lu, Foshan, 528000 China
| | - Huadong Zhou
- Maternity and Child Healthcare Hospital, Foshan Women and Children, 11 W. Renmin Lu, Foshan, 528000 China
| | - Weina Ju
- grid.420001.70000 0000 9813 9625New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Jerzy Wegiel
- grid.420001.70000 0000 9813 9625New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| | - Leslie Myatt
- grid.5288.70000 0000 9758 5690Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239 USA
| | - James M. Roberts
- grid.5288.70000 0000 9758 5690Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland, OR 97239 USA ,grid.460217.60000 0004 0387 4432Department of Obstetrics, Gynecology and Reproductive Sciences, Epidemiology and Clinical and Translational Research University of Pittsburgh, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, PA 15213 USA
| | - Xiaoling Guo
- Maternity and Child Healthcare Hospital, Foshan Women and Children, 11 W. Renmin Lu, Foshan, 528000 China
| | - Nanbert Zhong
- grid.420001.70000 0000 9813 9625New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314 USA
| |
Collapse
|
183
|
Hayder H, Shan Y, Chen Y, O’Brien JA, Peng C. Role of microRNAs in trophoblast invasion and spiral artery remodeling: Implications for preeclampsia. Front Cell Dev Biol 2022; 10:995462. [PMID: 36263015 PMCID: PMC9575991 DOI: 10.3389/fcell.2022.995462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
It is now well-established that microRNAs (miRNAs) are important regulators of gene expression. The role of miRNAs in placental development and trophoblast function is constantly expanding. Trophoblast invasion and their ability to remodel uterine spiral arteries are essential for proper placental development and successful pregnancy outcome. Many miRNAs are reported to be dysregulated in pregnancy complications, especially preeclampsia and they exert various regulatory effects on trophoblasts. In this review, we provide a brief overview of miRNA biogenesis and their mechanism of action, as well as of trophoblasts differentiation, invasion and spiral artery remodeling. We then discuss the role of miRNAs in trophoblasts invasion and spiral artery remodeling, focusing on miRNAs that have been thoroughly investigated, especially using multiple model systems. We also discuss the potential role of miRNAs in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Heyam Hayder
- Department of Biology, York University, Toronto, ON, Canada
| | - Yanan Shan
- Department of Biology, York University, Toronto, ON, Canada
| | - Yan Chen
- Department of Biology, York University, Toronto, ON, Canada
| | | | - Chun Peng
- Department of Biology, York University, Toronto, ON, Canada
- Centre for Research on Biomolecular Interactions, York University, Toronto, ON, Canada
- *Correspondence: Chun Peng,
| |
Collapse
|
184
|
Ninke T, Eifer A, Dieterich HJ. [Fetal and pediatric cardiovascular physiology : Things you should know as an (pediatric) anesthesiologist]. DIE ANAESTHESIOLOGIE 2022; 71:811-820. [PMID: 36053299 DOI: 10.1007/s00101-022-01198-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/25/2022] [Indexed: 06/15/2023]
Abstract
Immediately after birth the physiology of the cardiovascular system of the neonate undergoes some significant changes. The first breaths in life and the inflation of the lungs lead to a considerable drop in pulmonary arterial resistance. This results in the closure of the foramen ovale and ductus arteriosus; however, during the first weeks of life a sharp rise in pulmonary vascular resistance caused by hypoxia, hypercapnia and excessive positive pressure ventilation can lead to the reopening of the ductus arteriosus. This may result in subsequent strain of the left heart. In order to anticipate the reopening of the ductus arteriosus, it is recommended to measure the saturation of peripheral oxygen not only preductal (right hand), but also postductal (feet).An excessive volume therapy should be avoided as the neonatal myocardium is hallmarked by low cardiac compliance, reduced contractility and reduced ventricular filling.Until now there is still no uniform definition of hypotension in pediatric patients. Blood pressure values that are measured in awake children or are derived from the 50% age percentile values can thus only be used as approximate values. In all cases it is mandatory to recognize and consistently treat hypotension during pediatric anesthesia in order to prevent postoperative organ damage, particularly of the brain.The transcranial measurement of cerebral regional oxygen saturation (c‑rSO2) by means of near-infrared spectroscopy (NIRS) provides valuable information about regional tissue oxygenation of the brain. This enables conclusions about the state of the multifactorial cerebral perfusion to be drawn. In this way monitoring of the hypoxia sensitive cerebral tissue can be accomplished and should be used in premature infants and neonates. When measuring a baseline in awake patients, a 20% drop of c‑rSO2 from this baseline should be challenged and treated if necessary.
Collapse
Affiliation(s)
- T Ninke
- Klinik für Anaesthesiologie, Klinikum Universität München, Campus Innenstadt, Lindwurmstraße 2a, 80377, München, Deutschland.
| | - A Eifer
- Klinik für Anaesthesiologie, Klinikum Universität München, Campus Innenstadt, Lindwurmstraße 2a, 80377, München, Deutschland
| | - H-J Dieterich
- Klinik für Anaesthesiologie, Klinikum Universität München, Campus Innenstadt, Lindwurmstraße 2a, 80377, München, Deutschland
| |
Collapse
|
185
|
Upregulation of miR-181a-5p and miR-125b-2-3p in the Maternal Circulation of Fetuses with Rh-Negative Hemolytic Disease of the Fetus and Newborn Could Be Related to Dysfunction of Placental Function. DISEASE MARKERS 2022; 2022:2594091. [PMID: 36188428 PMCID: PMC9519318 DOI: 10.1155/2022/2594091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 08/21/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022]
Abstract
The transplacental transfer of maternal antibodies to the fetus is a critical mechanism for infant protection and perinatal disease. Hemolytic disease of the fetus and newborn (HDFN) is a representative fetal disease caused by transplacental transfer of maternal IgG antibodies. However, it is unclear whether placental-related miRNAs are expressed in Rh-HDFN. Through the investigation of the miR-181a-5p and miR-125b-2-3p levels in maternal plasma using qPCR, we found that both miR-181a-5p and miR-125b-2-3p were highly expressed in maternal plasma of newborns with Rh-HDFN compared with healthy controls, indicating the potential roles of these two miRNAs in Rh-HDFN. To demonstrate whether dysregulation of miR-125b-2-3p and miR-181a-5p contributes to Rh-HDFN development, we analyze the placental miRNA-/mRNA sequencing data (GSE73714) using weighted gene coexpression network analysis (WGCNA), miRNA target predictive databases, and DAVID (Database for Annotation, Visualization, and Integrated Discovery). The results showed that miR-125b-2-3p and miR-181a-5p could regulate several biological processes including cytoplasmic microtubule organization and angiogenesis. Moreover, core promoter sequence-specific DNA binding and protein binding were highly enriched molecular functions, indicating the potential roles of transcriptional regulation. Further pathway enrichment showed that miR-181a-5p and miR-125b-2-3p could regulate several biological pathways that were closely related to placental function, including the FoxO signaling pathway, focal adhesion, mTOR signaling pathway, and central carbon metabolism in cancer. In conclusion, the present results first revealed miRNA expression in the maternal circulation of newborns with Rh-HDFN, which could be caused by dysfunction of the placenta.
Collapse
|
186
|
Albraik RK, Shatla E, Abdulla YM, Ahmed EH. Neonatal Feeding Intolerance and Its Characteristics: A Descriptive Study. Cureus 2022; 14:e29291. [PMID: 36277537 PMCID: PMC9578381 DOI: 10.7759/cureus.29291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2022] [Indexed: 11/05/2022] Open
|
187
|
P06-21 MEHP treatment induces a decrease in SIRT1 expression, mitochondrial dysfunction, and transcriptomic alterations in the human trophoblastic cell line HTR-8/svneo. Toxicol Lett 2022. [DOI: 10.1016/j.toxlet.2022.07.339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
188
|
Cheng W, Zhang L, Sa P, Luo J, Li M. Transcriptomic analysis reveals the effects of maternal selenium deficiency on placental transport, hormone synthesis, and immune response in mice. METALLOMICS : INTEGRATED BIOMETAL SCIENCE 2022; 14:6674774. [PMID: 36002020 DOI: 10.1093/mtomcs/mfac062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 08/19/2022] [Indexed: 11/14/2022]
Abstract
Selenium deficiency has been considered to increase the risk of gestational complications. Our previous work showed that maternal selenium deficiency suppressed proliferation, induced autophagy dysfunction and apoptosis in the placenta of mice. However, other effects of maternal selenium deficiency on the placenta and the underlying mechanisms remain unclear. In the present study, dietary selenium deficiency in dams significantly suppressed glutathione peroxidase (GSH-Px) activity, total antioxidant capacity (T-AOC), and increased malondialdehyde (MDA) content in the placentae, confirming the oxidative stress in the placenta. By transcriptome sequencing analysis, the DEGs were involved in many biological processes, including ion transport, lipid metabolic process, immune response, transmembrane transport, and others. According to the KEGG analysis, the DEGs were primarily enriched in metabolic pathways, PI3K-Akt signaling pathway, and others. Among these, the steroid hormone biosynthesis pathway enriched the most DEGs. Hsd3b1, an ER enzyme involved in progesterone synthesis, was validated downregulated. Consistently, the progesterone content in the serum of the selenium-deficient group was decreased. Ion transporters and transmembrane transporters, such as Heph, Trf, Slc39a8, Slc23a1, Atp7b, and Kcnc1, were reduced in the selenium-deficient placentae. Immune response-related genes, including Ccl3, Ccl8, Cxcl10, and Cxcl14, were increased in the selenium-deficient placentae, along with an increase in macrophage number. These results suggested that maternal selenium deficiency may impair progesterone biosynthesis, reduce nutrient transporters expression, and promote immune response by increasing the oxidative stress of the placentae. This present study provides a novel insight into the possible cause of placenta disorder during pregnancy.
Collapse
Affiliation(s)
- Wanpeng Cheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Lantian Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Peiyue Sa
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Jing Luo
- Department of Clinical Medicine, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Mengdi Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.,Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| |
Collapse
|
189
|
Spathakis M, Filidou E, Pappa C, Arzou BC, Georgiadis A, Kontomanolis EN, Nikolettos N, Kolios G, Galazios G, Arvanitidis K. Spontaneous abortion is associated with differentially expressed angiogenic chemokines in placenta and decidua. Arch Gynecol Obstet 2022:10.1007/s00404-022-06725-8. [PMID: 35997970 DOI: 10.1007/s00404-022-06725-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/25/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Miscarriage is one of the most common complications of pregnancy. Although chromosomal abnormalities of the embryo is a well-known cause of miscarriage, a lot of cases remain unexplained, with immunologic and vascular growth alterations being considered as probable causes. Chemokines are produced by a variety of cells and exhibit several functions including both pro and anti-angiogenic properties. In this study, we investigated the role of the angiogenic and angiostatic chemokines in placenta and decidua tissues from spontaneous and induced abortions. METHODS Total RNA was extracted from the placenta and decidua tissues, which was then purified and converted into cDNA. Real-time PCR was then performed for the expression of the angiogenic CCL2, CCL5, CCL20, CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL7, CXCL8 and CXCL4, and the angiostatic CXCL9, CXCL10, CXCL11, CXCL12 and CXCL14 and results were then statistically analyzed. RESULTS Regarding the placenta, CXCL7 (2.29-fold, 2.16-2.38, p < 0.05), CXCL4 (1.01-fold, 0.74-4.447, p < 0.05), CXCL9 (0.87-fold, 0.43-1.34, p < 0.05) and CXCL11 (0.31-fold, 0.22-0.45, p < 0.05) were altered in spontaneous abortions. CCL2, CCL5, CXCL2-3, CXCL8, CXCL10, CXCL12 and CXCL14 were not statistically significant altered. Regarding the decidua, CXCL7 (7.13-fold, 6.32-7.54, p < 0.01), CXCL8 (11.02-fold, 8.58-13.45, p < 0.05), CCL20 (1.21-fold, 0.29-1.89, p < 0.05) and CXCL9 (5.49-fold, 3.67-6.39, p < 0.05) were overexpressed in spontaneous abortions. CXCL2-4, CCL2, CCL5, CXCL10-12 and CXCL14 did not show any differences. The expression of the chemokines CXCL1, CXCL5-6 was absent in either tissue or group. CONCLUSION Our results show that the overexpression of angiostatic and diminished expression of angiogenic chemokines takes place in the placenta and decidua of spontaneous abortions, suggesting that dysregulation of angiogenesis could be a contributive factor to the pathogenesis of miscarriage.
Collapse
Affiliation(s)
- Michail Spathakis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Chrysoula Pappa
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Bourazan Chalil Arzou
- Department of Obstetrics and Gynecology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Anastasios Georgiadis
- Department of Obstetrics and Gynecology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Emmanuel N Kontomanolis
- Department of Obstetrics and Gynecology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Nikos Nikolettos
- Department of Obstetrics and Gynecology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - George Galazios
- Department of Obstetrics and Gynecology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece.
| | - Konstantinos Arvanitidis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
190
|
Expression of ABCA1 Transporter and LXRA/LXRB Receptors in Placenta of Women with Late Onset Preeclampsia. J Clin Med 2022; 11:jcm11164809. [PMID: 36013052 PMCID: PMC9410380 DOI: 10.3390/jcm11164809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Appropriate levels of cholesterol are necessary for the mother and developing fetus, but theirexcess may cause preeclampsia. The ABCA1 transporter mediates the secretion of cholesterol and is highly regulated at the transcriptional level via the nuclear liver X receptors (LXRs). Methods: Sixteen preeclamptic and 39 normotensives healthy women with uncomplicated pregnancies were involved in the case-control study. The placental levels of ABCA1, LXRA and LXRB mRNA were quantified by real-time quantitative PCR. The concentrations of ABCA1, LXRA and LXRB proteins from the placenta were determined using an enzyme-linked immunosorbent assay Results: We found in the logistic regression model significantly lower placental expression of LXRB mRNA (crude OR = 0.26, 95% CI: 0.07–0.94, p = 0.040) and LXRA protein level (crude OR = 0.19, 95% CI: 0.05–0.69, p = 0.012) in late-onset preeclamptic women compared to healthy pregnant women. The values remained statistically significant after adjustment for possible confounders. Conclusions: Our results suggest that high placenta LXRA mRNA and LXRA protein expression levels decrease the risk of late-onset preeclampsia. These nuclear receptors could play a role in the development of preeclampsia through disturbances of lipid metabolism.
Collapse
|
191
|
Kim SY, Lee YJ, An SM, Kim MJ, Jeong JS, Kim DS, Lim Y, Jung EM, Kim SC, An BS. Dynamic regulation of lipid metabolism in the placenta of in vitro and in vivo models of Gestational Diabetes Mellitus. Biol Reprod 2022; 107:1311-1318. [PMID: 35932454 DOI: 10.1093/biolre/ioac156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 07/22/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
The purpose of this study was to investigate lipid metabolism in the placenta of Gestational diabetes mellitus (GDM) individuals and to evaluate its effect on the fetus. We examined the expression of lipogenesis- and lipolysis-related proteins in the in vitro and in vivo GDM placenta models. The levels of sterol regulatory element binding protein-1c (SREBP-1c) were increased, and fat accumulated more during early hyperglycemia, indicating that lipogenesis was stimulated. When hyperglycemia was further extended, lipolysis was activated due to the phosphorylation of hormone-sensitive lipase (HSL) and expression of adipose triglyceride lipase (ATGL). In the animal model of GDM and in the placenta of GDM patients during the extended stage of GDM, the expression of SREBP-1c decreased and the deposition of fat increased. Similar to the results obtained in the in vitro study, lipolysis was enhanced in the animal and human placenta of extended GDM. These results suggest that fat synthesis may be stimulated by lipogenesis in the placenta when the blood glucose level is high. Subsequently, the accumulated fat can be degraded by lipolysis and more fat and its metabolites can be delivered to the fetus when the GDM condition is extended at the late stage of gestation. Imbalanced fat metabolism in the placenta and fetus of GDM patients can cause metabolic complications in the fetus, including fetal macrosomia, obesity, and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- So Young Kim
- Department of Biomaterials Science (BK21 Four Program), College of Natural Resources & Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Young Joo Lee
- Department of Obstetrics and Gynecology, School of Medicine, Kyung Hee Medical Center, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sung-Min An
- Department of Biomaterials Science (BK21 Four Program), College of Natural Resources & Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Min Jae Kim
- Department of Biomaterials Science (BK21 Four Program), College of Natural Resources & Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Jea Sic Jeong
- Department of Biomaterials Science (BK21 Four Program), College of Natural Resources & Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Da Som Kim
- Department of Biomaterials Science (BK21 Four Program), College of Natural Resources & Life Science, Pusan National University, Miryang 50463, Republic of Korea
| | - Yong Lim
- Department of Clinical Laboratory Science, College of Nursing and Healthcare Science, Dong-Eui University, Busan, Republic of Korea
| | - Eui-Man Jung
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Seung-Chul Kim
- Department of Obstetrics and Gynecology, Biomedical Research Institute, Pusan National University School of Medicine, Busan, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterials Science (BK21 Four Program), College of Natural Resources & Life Science, Pusan National University, Miryang 50463, Republic of Korea
| |
Collapse
|
192
|
Tamayo JM, Rose D, Church JS, Schwartzer JJ, Ashwood P. Maternal Allergic Asthma Induces Prenatal Neuroinflammation. Brain Sci 2022; 12:brainsci12081041. [PMID: 36009104 PMCID: PMC9405898 DOI: 10.3390/brainsci12081041] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Autism spectrum disorder (ASD) is a class of neurodevelopmental disorders characterized by impaired social interactions and communication skills and repetitive or stereotyped behaviors. Rates of ASD diagnosis continue to rise, with current estimates at 1 in 44 children in the US (Maenner 2021). Epidemiological studies have suggested a link between maternal allergic asthma and an increased likelihood of having a child diagnosed with ASD. However, a lack of robust laboratory models prevents mechanistic research from being carried out. We developed a novel mouse model of maternal asthma-allergy (MAA) and previously reported that offspring from these mothers exhibit behavioral deficits compared to controls. In addition, it was shown that epigenetic regulation of gene expression in microglia was altered in these offspring, including several autism candidate genes. To further elucidate if there is neuroinflammation in the fetus following MAA, we investigated how allergic asthma impacts the maternal environment and inflammatory markers in the placenta and fetal brain during gestation. Female C57Bl/6 mice were primed with ovalbumin (OVA) prior to allergic asthma induction during pregnancy by administering aerosolized ovalbumin or PBS control to pregnant dams at gestational days (GD)9.5, 12.5, and 17.5. Four hours after the final induction, placenta and fetal brains were collected and measured for changes in cytokines using a Luminex bead-based multiplex assay. Placental MAA tissue showed a decrease in interleukin (IL)-17 in male and female offspring. There was a sex-dependent decrease in female monocyte chemoattractant protein 1 (MCP-1). In male placentas, IL-4, C-X-C motif chemokine 10 (CXCL10)-also known as interferon γ-induced protein 10 kDa (IP-10)-and chemokine (C-C motif) ligand 5 (RANTES) were decreased. In fetal brains, elevated inflammatory cytokines were found in MAA offspring when compared to controls. Specifically, interferon-gamma (IFN-γ), granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin 1α (IL-1α), IL-6, and tumor necrosis factor α (TNFα) were elevated in both males and females. In contrast, a decrease in the cytokine IL-9 was also observed. There were slight sex differences after OVA exposures. Male fetal brains showed elevated levels of macrophage inflammatory protein-2 (MIP-2), whereas female brains showed increased keratinocytes-derived chemokine (KC). In addition, IL-1𝛽 and IP-10 in male fetal brains were decreased. Together, these data indicate that repeated exposure to allergic asthma during pregnancy alters cytokine expression in the fetal environment in a sex-dependent way, resulting in homeostatic and neuroinflammatory alterations in the fetal brain.
Collapse
Affiliation(s)
- Juan M. Tamayo
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95817, USA
- The M.I.N.D. Institute, University of California Davis, Sacramento, CA 95817, USA
| | - Destanie Rose
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95817, USA
- The M.I.N.D. Institute, University of California Davis, Sacramento, CA 95817, USA
| | - Jamie S. Church
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Jared J. Schwartzer
- Program in Neuroscience and Behavior, Department of Psychology and Education, Mount Holyoke College, 50 College Street, South Hadley, MA 01075, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95817, USA
- The M.I.N.D. Institute, University of California Davis, Sacramento, CA 95817, USA
- Correspondence:
| |
Collapse
|
193
|
Grant AD, Erickson EN. Birth, love, and fear: Physiological networks from pregnancy to parenthood. COMPREHENSIVE PSYCHONEUROENDOCRINOLOGY 2022; 11:100138. [PMID: 35757173 PMCID: PMC9227990 DOI: 10.1016/j.cpnec.2022.100138] [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: 11/29/2021] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 12/01/2022] Open
Abstract
Pregnancy and childbirth are among the most dramatic physiological and emotional transformations of a lifetime. Despite their central importance to human survival, many gaps remain in our understanding of the temporal progression of and mechanisms underlying the transition to new parenthood. The goal of this paper is to outline the physiological and emotional development of the maternal-infant dyad from late pregnancy to the postpartum period, and to provide a framework to investigate this development using non-invasive timeseries. We focus on the interaction among neuroendocrine, emotional, and autonomic outputs in the context of late pregnancy, parturition, and post-partum. We then propose that coupled dynamics in these outputs can be leveraged to map both physiologic and pathologic pregnancy, parturition, and parenthood. This approach could address gaps in our knowledge and enable early detection or prediction of problems, with both personalized depth and broad population scale.
Collapse
Affiliation(s)
- Azure D. Grant
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, United States
- Levels Health Inc., 228 Park Ave. South, PMB 63877, New York, NY, 10003, United States
| | - Elise N. Erickson
- Oregon Health and Science University, Portland, OR, 97239, United States
| |
Collapse
|
194
|
Placental dysfunction: The core mechanism for poor neurodevelopmental outcomes in the offspring of preeclampsia pregnancies. Placenta 2022; 126:224-232. [PMID: 35872512 DOI: 10.1016/j.placenta.2022.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022]
Abstract
Preeclampsia (PE) is a leading condition threatening pregnant women and their offspring. The offspring of PE pregnancies have a high risk of poor neurodevelopmental outcomes and neuropsychological diseases later in life. However, the pathophysiology and pathogenesis of poor neurodevelopment remain undetermined. Abnormal placental functions are at the core of most PE cases, and recent research evidence supports that the placenta plays an important role in fetal brain development. Here, we summarize the relationship between abnormal fetal brain development and placental dysfunction in PE conditions, which include the dysfunction of nutrient and gas-waste exchange, impaired angiogenesis stimulation, abnormal neurotransmitter regulation, disrupted special protectors, and immune disorders. All these factors could lead to poor neurodevelopmental outcomes.
Collapse
|
195
|
Transforming growth factor-β signaling governs the differentiation program of extravillous trophoblasts in the developing human placenta. Proc Natl Acad Sci U S A 2022; 119:e2120667119. [PMID: 35867736 PMCID: PMC9282384 DOI: 10.1073/pnas.2120667119] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Abnormal placentation has been noticed in a variety of pregnancy complications such as miscarriage, early-onset preeclampsia, and fetal growth restriction. Defects in the developmental program of extravillous trophoblasts (EVTs), migrating from placental anchoring villi into the maternal decidua and its vessels, is thought to be an underlying cause. Yet, key regulatory mechanisms controlling commitment and differentiation of the invasive trophoblast lineage remain largely elusive. Herein, comparative gene expression analyses of HLA-G-purified EVTs, isolated from donor-matched placenta, decidua, and trophoblast organoids (TB-ORGs), revealed biological processes and signaling pathways governing EVT development. In particular, bioinformatics analyses and manipulations in different versatile trophoblast cell models unraveled transforming growth factor-β (TGF-β) signaling as a crucial pathway driving differentiation of placental EVTs into decidual EVTs, the latter showing enrichment of a secretory gene signature. Removal of Wingless signaling and subsequent activation of the TGF-β pathway were required for the formation of human leukocyte antigen-G+ (HLA-G+) EVTs in TB-ORGs that resemble in situ EVTs at the level of global gene expression. Accordingly, TGF-β-treated EVTs secreted enzymes, such as DAO and PAPPA2, which were predominantly expressed by decidual EVTs. Their genes were controlled by EVT-specific induction and genomic binding of the TGF-β downstream effector SMAD3. In summary, TGF-β signaling plays a key role in human placental development governing the differentiation program of EVTs.
Collapse
|
196
|
Koshko L, Scofield S, Mor G, Sadagurski M. Prenatal Pollutant Exposures and Hypothalamic Development: Early Life Disruption of Metabolic Programming. Front Endocrinol (Lausanne) 2022; 13:938094. [PMID: 35909533 PMCID: PMC9327615 DOI: 10.3389/fendo.2022.938094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Environmental contaminants in ambient air pollution pose a serious risk to long-term metabolic health. Strong evidence shows that prenatal exposure to pollutants can significantly increase the risk of Type II Diabetes (T2DM) in children and all ethnicities, even without the prevalence of obesity. The central nervous system (CNS) is critical in regulating whole-body metabolism. Within the CNS, the hypothalamus lies at the intersection of the neuroendocrine and autonomic systems and is primarily responsible for the regulation of energy homeostasis and satiety signals. The hypothalamus is particularly sensitive to insults during early neurodevelopmental periods and may be susceptible to alterations in the formation of neural metabolic circuitry. Although the precise molecular mechanism is not yet defined, alterations in hypothalamic developmental circuits may represent a leading cause of impaired metabolic programming. In this review, we present the current knowledge on the links between prenatal pollutant exposure and the hypothalamic programming of metabolism.
Collapse
Affiliation(s)
- Lisa Koshko
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Sydney Scofield
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology School of Medicine, Wayne State University, Detroit, MI, United States
| | - Marianna Sadagurski
- Integrative Biosciences Center, Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| |
Collapse
|
197
|
Ruyak SL, Noor S, DiDomenico J, Sun MS, Fernandez Oropeza AK, Rodriguez DE, Marquez LE, Milligan ED, Bakhireva LN. Effects of prenatal opioid and alcohol exposures on immune and serotonin factors in human placenta. Exp Neurol 2022; 353:114057. [PMID: 35364108 PMCID: PMC10035581 DOI: 10.1016/j.expneurol.2022.114057] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Opioids and alcohol impact critical serotonin (5-HT) function in the developing placenta and fetus through the actions of immune proinflammatory factors. Yet, possible convergent effects of opioids and alcohol on human placental toll-like receptor 4 (TLR4) activation and subsequent 5-HT homeostasis remain entirely unknown. The purpose of this study was to examine the effect of prenatal exposure to opioids with or without prenatal alcohol exposure (PAE) on the expression of key placental immune and serotonin signaling factors in human placental tissue obtained from a well-characterized prospective cohort. METHODS Data were collected from a subset of participants enrolled in the prospective pre-birth Ethanol, Neurodevelopment, Infant, and Child Health (ENRICH-1) cohort. Women were recruited and classified into four study groups: 1) PAE (n = 20); 2) those taking medications for opioid use disorder (MOUD; n = 28), 3) concurrent PAE and MOUD (n = 20); and 4) controls (HC; n = 20) based on prospective, repeated self-report, and biomarker analysis. Placenta samples underwent tissue processing to identify mRNA for TLR4, nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), serotonin transporter (SERT), tryptophan hydroxylase (TPH1), indoleamine 2,3-Dioxygenase 1 (IDO) as well as protein concentrations of TLR4, IL-1β, TNF-α, SERT. To consider the association between study group and mRNA/protein expression of our targets, multivariable regression models were developed with inclusion of a priori selected covariates. RESULTS There was a significant negative association between PAE and SERT mRNA (β = -0.01; p < 0.01) and a positive association with TPH1 mRNA expression (β = 0.78; p < 0.05). In addition, there was a negative association between MOUD and TNF-α protein expression (β = -0.12; p < 0.05). CONCLUSIONS This study provides the first evidence that PAE may inhibit SERT expression while simultaneously promoting increased TPH1 protein expression in human placenta. This may result in increased 5-HT in fetal circulation known to affect neurodevelopment. Our data suggest that opioids and alcohol may disturb the bidirectional, dynamic interaction between the placental immune and serotonin system. Given the implication for brain development and health across the life-span further investigation of these critical mechanisms in well-defined cohorts is required.
Collapse
Affiliation(s)
- Sharon L Ruyak
- College of Nursing, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America; College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America.
| | - Shahani Noor
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States of America
| | - Jared DiDomenico
- College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Melody S Sun
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States of America
| | - Annette K Fernandez Oropeza
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States of America
| | - Dominique E Rodriguez
- College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Lidia Enriquez Marquez
- College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| | - Erin D Milligan
- Department of Neurosciences, University of New Mexico, Health Sciences Center, Albuquerque, NM, United States of America
| | - Ludmila N Bakhireva
- College of Pharmacy Substance Use Research Education Center, University of New Mexico Health Sciences Center, Albuquerque, NM, United States of America
| |
Collapse
|
198
|
Ramos IF, Ross KM, Rinne GR, Somers JA, Mancuso RA, Hobel CJ, Coussons-Read M, Dunkel Schetter C. Pregnancy anxiety, placental corticotropin-releasing hormone and length of gestation. Biol Psychol 2022; 172:108376. [PMID: 35667479 PMCID: PMC10022399 DOI: 10.1016/j.biopsycho.2022.108376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/20/2022]
Abstract
OBJECTIVE High pregnancy anxiety is a consistent predictor of earlier labor and delivery. Placental corticotropin-releasing hormone (pCRH) predicts earlier delivery consistently and it has been identified as a biological mediator of the association between pregnancy anxiety and gestational length. However, studies have not examined whether changes in pregnancy anxiety are associated with earlier birth as mediated by changes in pCRH during pregnancy. Accordingly, this study tests whether linear changes in pregnancy anxiety are associated with length of gestation indirectly through nonlinear increases in pCRH over pregnancy. METHODS A sample of pregnant women (n=233) completed prenatal assessments in early pregnancy, second trimester, and third trimester that included a 4-item assessment of pregnancy anxiety and collection of blood samples assayed for pCRH using radioimmunoassay. Length of gestation was abstracted from medical records after birth. RESULTS Increases in pregnancy anxiety from early pregnancy to third trimester predicted shorted length of gestation, as did nonlinear increases in pCRH over pregnancy. However, there was no evidence of an indirect effect of changes in pregnancy anxiety on length of gestation via changes in pCRH. CONCLUSIONS These results indicate that linear changes in pregnancy anxiety and nonlinear changes in pCRH during pregnancy are independent risk factors for shortened gestational length. This study adds to a small but growing body of work on biopsychological processes in pregnancy and length of gestation. Modeling changes in psychological and biological processes during pregnancy could provide more insight into understanding risk for adverse pregnancy outcomes.
Collapse
|
199
|
Zhang Y, Zhong Y, Zou L, Liu X. Significance of Placental Mesenchymal Stem Cell in Placenta Development and Implications for Preeclampsia. Front Pharmacol 2022; 13:896531. [PMID: 35721156 PMCID: PMC9198303 DOI: 10.3389/fphar.2022.896531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/16/2022] [Indexed: 12/29/2022] Open
Abstract
The well-developed placentation is fundamental for the reproductive pregnancy while the defective placental development is the pathogenetic basis of preeclampsia (PE), a dangerous complication of pregnancy comprising the leading causes of maternal and perinatal morbidity and mortality. Placenta-derived mesenchymal stem cells (PMSCs) are a group of multipotent stem cells that own a potent capacity of differentiating into constitutive cells of vessel walls. Additionally, with the paracrine secretion of various factors, PMSCs inextricably link and interact with other component cells in the placenta, collectively improving the placental vasculature, uterine spiral artery remolding, and uteroplacental interface immunoregulation. Recent studies have further indicated that preeclamptic PMSCs, closely implicated in the abnormal crosstalk between other ambient cells, disturb the homeostasis and development in the placenta. Nevertheless, PMSCs transplantation or PMSCs exosome therapies tend to improve the placental vascular network and trophoblastic functions in the PE model, suggesting PMSCs may be a novel and putative therapeutic strategy for PE. Herein, we provide an overview of the multifaceted contributions of PMSCs in early placental development. Thereinto, the intensive interactions between PMSCs and other component cells in the placenta were particularly highlighted and further extended to the implications in the pathogenesis and therapeutic strategies of PE.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqi Zhong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
200
|
Berger N, Allerkamp H, Wadsack C. Serine Hydrolases in Lipid Homeostasis of the Placenta-Targets for Placental Function? Int J Mol Sci 2022; 23:6851. [PMID: 35743292 PMCID: PMC9223866 DOI: 10.3390/ijms23126851] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 02/01/2023] Open
Abstract
The metabolic state of pregnant women and their unborn children changes throughout pregnancy and adapts to the specific needs of each gestational week. These adaptions are accomplished by the actions of enzymes, which regulate the occurrence of their endogenous substrates and products in all three compartments: mother, placenta and the unborn. These enzymes determine bioactive lipid signaling, supply, and storage through the generation or degradation of lipids and fatty acids, respectively. This review focuses on the role of lipid-metabolizing serine hydrolases during normal pregnancy and in pregnancy-associated pathologies, such as preeclampsia, gestational diabetes mellitus, or preterm birth. The biochemical properties of each class of lipid hydrolases are presented, with special emphasis on their role in placental function or dysfunction. While, during a normal pregnancy, an appropriate tonus of bioactive lipids prevails, dysregulation and aberrant signaling occur in diseased states. A better understanding of the dynamics of serine hydrolases across gestation and their involvement in placental lipid homeostasis under physiological and pathophysiological conditions will help to identify new targets for placental function in the future.
Collapse
Affiliation(s)
- Natascha Berger
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (N.B.); (H.A.)
| | - Hanna Allerkamp
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (N.B.); (H.A.)
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria; (N.B.); (H.A.)
- BioTechMed-Graz, 8036 Graz, Austria
| |
Collapse
|