1
|
Anderson EC, Muse ME, Niu Z, Foley HB, Levy JJ, Romano ME, Gui J, Bentz JL, Farzan SF, Bastain TM, Marsit CJ, Breton CV, Howe CG. Blood Pressure and Late Pregnancy Circulating miRNAs in the MADRES Study. J Am Heart Assoc 2025; 14:e040416. [PMID: 40497511 DOI: 10.1161/jaha.124.040416] [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: 12/02/2024] [Accepted: 04/24/2025] [Indexed: 06/19/2025]
Abstract
BACKGROUND Circulating extracellular and vesicle particle (EVP) miRNAs have been associated with cardiovascular risk and adverse birth outcomes. Hypertensive disorders of pregnancy (HDP) increase risk for adverse birth outcomes and future cardiovascular outcomes in mothers and children and have been associated with altered maternal circulating EVP miRNA levels during pregnancy. Whether these relationships exist for elevated blood pressure (BP) in the subclinical range is unknown. We investigated associations between (1) hypertensive disorders of pregnancy and (2) maternal BP trajectories, including in the subclinical range, and circulating EVP miRNA levels during pregnancy in the MADRES (Maternal and Developmental Risks From Environmental and Social Stressors) Study (n=372). METHODS Latent class trajectory modeling was used to identify trajectories from BP measures abstracted from medical records. The NanoString nCounter platform was used to quantify 798 miRNAs extracted from maternal blood (median gestational age: 31.6 weeks). Covariate-adjusted regression models assessed associations between each hypertensive disorders of pregnancy subtype or BP trajectory and levels of each miRNA. RESULTS Three BP trajectories were identified: Low, Moderate, and High. Chronic hypertension was associated with higher levels of miR-1185-2-3p (Pfalse discovery rate<0.05), a placenta-specific miRNA linked to arterial stiffness and preterm delivery. Many placenta-expressed miRNAs previously associated with a longer gestational duration in the same cohort were lower among participants with elevated BP (P<0.05). Target genes of BP-associated EVP miRNAs were overrepresented in pathways involved in vascular inflammation, oxidative stress, endothelial dysfunction, and placental function. CONCLUSIONS Circulating levels of placenta-expressed EVP miRNAs previously implicated in adverse birth and cardiovascular outcomes are sensitive to elevated maternal BP during pregnancy, including in the subclinical range.
Collapse
Affiliation(s)
| | - Meghan E Muse
- Department of Epidemiology Geisel School of Medicine at Dartmouth Lebanon NH
| | - Zhongzheng Niu
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine University of Southern California Los Angeles CA
| | - Helen B Foley
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine University of Southern California Los Angeles CA
| | - Joshua J Levy
- Emerging Diagnostic and Investigative Technologies, Department of Pathology and Laboratory Medicine Dartmouth Hitchcock Medical Center Lebanon NH
- Department of Pathology and Laboratory Medicine and the Department of Computational Biomedicine Cedars-Sinai Medical Center Los Angeles CA
| | - Megan E Romano
- Department of Epidemiology Geisel School of Medicine at Dartmouth Lebanon NH
| | - Jiang Gui
- Department of Biomedical Data Science Geisel School of Medicine at Dartmouth Hanover NH
| | - Jessica L Bentz
- Department of Pathology Dartmouth-Hitchcock Medical Center Lebanon NH
| | - Shohreh F Farzan
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine University of Southern California Los Angeles CA
| | - Theresa M Bastain
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine University of Southern California Los Angeles CA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Rollins School of Public Health Emory University Atlanta GA
| | - Carrie V Breton
- Division of Environmental Health, Department of Population and Public Health Sciences, Keck School of Medicine University of Southern California Los Angeles CA
| | - Caitlin G Howe
- Department of Epidemiology Geisel School of Medicine at Dartmouth Lebanon NH
| |
Collapse
|
2
|
Kosztowny E, Wawrzykowski JT, Jamioł MA, Kankofer M. The Profile of Selected Protein Markers of Senescence in the Placentas of Cows During Early-Mid-Pregnancy and Parturition with and Without the Retention of Fetal Membranes: A Preliminary Study. Int J Mol Sci 2025; 26:5475. [PMID: 40564952 DOI: 10.3390/ijms26125475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/25/2025] [Accepted: 06/05/2025] [Indexed: 06/28/2025] Open
Abstract
Senescence in placental cells impacts physiological functions and contributes to pathology. Therefore, we examined biochemical markers of cellular senescence-p38, P-p38, p21, and p53-during pregnancy, at parturition, and in cases of retained fetal membranes. Placentomes were collected from pregnant cows (2nd, 4th, and 5th months of gestation) and parturient cows undergoing cesarean section, categorized as NR or RFM. Samples were separated into maternal and fetal parts and analyzed via WB and ELISA. WB confirmed protein presence, while ELISA showed a significant increase in the concentrations of both p38 and P-p38 in the fetal part in the 5th month of gestation as compared to earlier months. No significant differences were observed in the maternal part across pregnancy and parturition. These findings suggest p38 and P-p38 may be important molecules regulating the normal development of the bovine placenta during middle pregnancy. Further studies are needed to elucidate the mechanisms of their action.
Collapse
Affiliation(s)
- Ewelina Kosztowny
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka Street 12, 20-033 Lublin, Poland
| | - Jacek T Wawrzykowski
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka Street 12, 20-033 Lublin, Poland
| | - Monika A Jamioł
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka Street 12, 20-033 Lublin, Poland
| | - Marta Kankofer
- Department of Biochemistry, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka Street 12, 20-033 Lublin, Poland
| |
Collapse
|
3
|
Fasoulopoulos A, Varras M, Varra FN, Philippou A, Myoteri D, Varra VK, Kouroglou E, Gryparis A, Papadopetraki A, Vlachos I, Papadopoulos K, Koutsilieris M, Konstantinidou AE. Expression of the IGF‑1Ea isoform in human placentas from third trimester normal and idiopathic intrauterine growth restriction singleton pregnancies: Correlations with clinical and histopathological parameters. Mol Med Rep 2025; 31:69. [PMID: 39791214 PMCID: PMC11751665 DOI: 10.3892/mmr.2025.13434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/17/2024] [Indexed: 01/12/2025] Open
Abstract
Intrauterine growth restriction (IUGR) is the second most common obstetric complication after preterm labor. Appropriate trophoblast differentiation and placental structure, growth and function are key for the maintenance of pregnancy and normal fetal growth, development and survival. Extravillous trophoblast cell proliferation, migration and invasion are regulated by molecules produced by the fetomaternal interface, including autocrine factors produced by the trophoblast, such as insulin‑like growth factor (IGF)‑1. The aim of the present study was to investigate expression patterns of IGF‑1Ea isoform in IUGR placenta compared with appropriate for gestational age (AGA) pregnancies. Placental frozen tissues were collected from 13 AGA and 15 IUGR third trimester pregnancies for detection of IGF‑1Ea mRNA expression using reverse transcription‑quantitative PCR. Formalin‑fixed paraffin‑embedded samples from 15 AGA and 47 IUGR pregnancies were analyzed immunohistochemically for the identification and localization of the IGF‑1Ea peptide and comparison of clinical and histopathological parameters. To the best of our knowledge, the present study is the first to show IGF‑1Ea expression in third trimester human placenta. The results indicated that similar IGF‑1Ea mRNA expression levels were present in placental specimens from both groups. Cytoplasmic IGF‑1Ea expression was localized in the perivillous syncytiotrophoblast, extravillous trophoblast and endothelium of the villous and decidual vessels in both groups. No significant difference in the scores and intensity of IGF‑1Ea expression in perivillous syncytiotrophoblasts were noted in the IUGR vs. AGA pregnancies. Most IUGR cases showed negative IGF‑1Ea expression in the extravillous trophoblast, whereas AGA pregnancies showed predominantly positive immunostaining. A sex‑specific expression pattern was noted in the extravillous trophoblast, with negative IGF‑1Ea expression in the placentas of female IUGR cases. Additionally, positive immunostaining for IGF‑1Ea peptide in fetal villous and maternal decidual vessels, was more frequently observed in the IUGR group compared with AGA. In conclusion, no difference in total IGF‑1Ea mRNA placental expression was observed between IUGR and AGA pregnancies, likely due to heterogeneity of histological structures expressing this isoform. Negative IGF‑1Ea immunohistological expression in the extravillous trophoblast from IUGR placentas, associated with histological changes of maternal malperfusion, may reflect the involvement of this isoform in defective placentation. The presence of IGF‑1Ea peptide in the endothelium of the villous vessels in IUGR placentas may indicate a reactive autocrine regulation to compensate for malperfused villi in IUGR pregnancy by regulating angiogenesis and vasodilation. The observed sex differences in IGF‑1Ea expression between IUGR and AGA placentas may indicate interactions between sex hormones and selective IGF‑1 binding proteins in regulating IGF‑1Ea synthesis; however, this requires further elucidation.
Collapse
Affiliation(s)
- Apostolos Fasoulopoulos
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Michail Varras
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Fani-Niki Varra
- Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Anastasios Philippou
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Despina Myoteri
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | | | - Evgenia Kouroglou
- Fourth Obstetrics and Gynecology Department, ‘Elena Venizelou’ General Hospital of Athens, 11521 Athens, Greece
| | - Alexandros Gryparis
- Department of Speech and Language Therapy, University of Ioannina, 45500 Ioannina, Greece
| | - Argyro Papadopetraki
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Iakovos Vlachos
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Konstantinos Papadopoulos
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National Kapodistrian University of Athens, 11527 Athens, Greece
| | - Anastasia Evangelia Konstantinidou
- Department of Pathology, Aretaieion University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
- First Department of Pathology, Unit of Pediatric-Perinatal Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
4
|
Murthi P, Kalionis B. Homeobox genes in the human placenta: Twists and turns on the path to find novel targets. Placenta 2024; 157:28-36. [PMID: 38908943 DOI: 10.1016/j.placenta.2024.06.010] [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: 03/16/2024] [Revised: 05/25/2024] [Accepted: 06/16/2024] [Indexed: 06/24/2024]
Abstract
Fetal growth restriction (FGR) is a clinically important human pregnancy disorder that is thought to originate early in pregnancy and while its aetiology is not well understood, the disorder is associated with placental insufficiency. Currently treatment for FGR is limited by increased surveillance using ultrasound monitoring and premature delivery, or corticosteroid medication in the third trimester to prolong pregnancy. There is a pressing need for novel strategies to detect and treat FGR at its early stage. Homeobox genes are well established as master regulators of early embryonic development and increasing evidence suggests they are also important in regulating early placental development. Most important is that specific homeobox genes are abnormally expressed in human FGR. This review focusses on identifying the molecular pathways controlled by homeobox genes in the normal and FGR-affected placenta. This information will begin to address the knowledge gap in the molecular aetiology of FGR and lay the foundation for identifying potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Padma Murthi
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women's Hospital and Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Victoria, Australia.
| | - Bill Kalionis
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women's Hospital and Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
5
|
de Dios N, Riedel R, Schanton M, Balestrini P, Pérez L, Pérez-Pérez A, Etcheverry T, Casale R, Farina M, Sánchez-Margalet V, Maymó J, Varone C. Placental apoptosis increased by hypoxia inducible factor-1 stabilization is counteracted by leptin†. Biol Reprod 2024; 111:708-722. [PMID: 38924703 DOI: 10.1093/biolre/ioae095] [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: 03/23/2024] [Revised: 05/16/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
During pregnancy, apoptosis is a physiological event critical in the remodeling and aging of the placenta. Increasing evidence has pointed toward the relevance of hypoxia as modulator of trophoblast cell death. Previous reports have shown that leptin, a placental cytokine, promotes cell survival in both cell culture and placental explant models. The aim of this work is to establish the role of leptin in apoptosis under hypoxic condition in trophoblast cells. In this study, we evaluated the effect of cobalt chloride, a hypoxia mimicking agent that stabilizes the expression of hypoxia-inducible factor-1 alpha, on Swan-71 and human placental explants. Hypoxia chamber was also used to generate 2% oxygen. Apoptosis was determined by the presence of apoptotic nucleus, fragmentation of DNA and Caspase-3 and PARP-1 cleavage. The pro-apoptotic proteins BAX, BID, BAD, and BAK and the anti-apoptotic effectors BCL-2, B-cell lymphoma-extra-large, and myeloid cell leukemia-1 were also analyzed. We found that hypoxia-inducible factor-1 alpha stabilization increased the appearance of apoptotic nucleus, fragmentation of DNA, and Caspase-3 and PARP-1 cleavage. Hypoxia mimicking conditions enhanced the expression of pro-apoptotic effectors BAX, BID, BAD, and BAK. Hypoxia-inducible factor-1 alpha stabilization also downregulated the level of BCL-2, B-cell lymphoma-extra-large, and myeloid cell leukemia-1. All these apoptotic parameters changes were reversed with leptin treatment. Moreover, we showed that leptin action on apoptosis modulation involves PI3K and MAPK signaling pathways. Obtained data demonstrate that hypoxia-inducible factor-1 alpha stabilization induces apoptosis in human placenta and leptin counteracts this effect, reinforcing its role as a survival cytokine.
Collapse
Affiliation(s)
- Nataly de Dios
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Rodrigo Riedel
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Malena Schanton
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Paula Balestrini
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Luciano Pérez
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Antonio Pérez-Pérez
- Departamento de Bioquímica Médica y Biología Molecular. Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Tomás Etcheverry
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO, CONICET), Universidad de Buenos Aires. Facultad de Medicina, Buenos Aires, Argentina
| | - Roberto Casale
- Departamento Materno-Infantil, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Mariana Farina
- Centro de Estudios Farmacológicos y Botánicos (CEFYBO, CONICET), Universidad de Buenos Aires. Facultad de Medicina, Buenos Aires, Argentina
| | - Víctor Sánchez-Margalet
- Departamento de Bioquímica Médica y Biología Molecular. Hospital Universitario Virgen Macarena, Facultad de Medicina, Universidad de Sevilla, Sevilla, España
| | - Julieta Maymó
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| | - Cecilia Varone
- Departamento de Química Biológica, Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Buenos Aires, Argentina
- Universidad de Buenos Aires. CONICET. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), Buenos Aires, Argentina
| |
Collapse
|
6
|
Suda M, Paul KH, Tripathi U, Minamino T, Tchkonia T, Kirkland JL. Targeting Cell Senescence and Senolytics: Novel Interventions for Age-Related Endocrine Dysfunction. Endocr Rev 2024; 45:655-675. [PMID: 38500373 PMCID: PMC11405506 DOI: 10.1210/endrev/bnae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 01/11/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Multiple changes occur in hormonal regulation with aging and across various endocrine organs. These changes are associated with multiple age-related disorders and diseases. A better understanding of responsible underling biological mechanisms could help in the management of multiple endocrine disorders over and above hormone replacement therapy (HRT). Cellular senescence is involved in multiple biological aging processes and pathologies common in elderly individuals. Cellular senescence, which occurs in many older individuals but also across the lifespan in association with tissue damage, acute and chronic diseases, certain drugs, and genetic syndromes, may contribute to such endocrine disorders as osteoporosis, metabolic syndrome, and type 2 diabetes mellitus. Drugs that selectively induce senescent cell removal, "senolytics,", and drugs that attenuate the tissue-destructive secretory state of certain senescent cells, "senomorphics," appear to delay the onset of or alleviate multiple diseases, including but not limited to endocrine disorders such as diabetes, complications of obesity, age-related osteoporosis, and cancers as well as atherosclerosis, chronic kidney disease, neurodegenerative disorders, and many others. More than 30 clinical trials of senolytic and senomorphic agents have already been completed, are underway, or are planned for a variety of indications. Targeting senescent cells is a novel strategy that is distinct from conventional therapies such as HRT, and thus might address unmet medical needs and can potentially amplify effects of established endocrine drug regimens, perhaps allowing for dose decreases and reducing side effects.
Collapse
Affiliation(s)
- Masayoshi Suda
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
| | - Karl H Paul
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Pharmacology, Karolinska Institutet, Solnavägen 9, 171 65 Solna, Sweden
| | - Utkarsh Tripathi
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutionary Medical Science and Technology (AMED-CREST), Japan Agency for Medical Research and Development, Tokyo, 100-0004, Japan
| | - Tamara Tchkonia
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Departments of Medicine and Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
- Division of General Internal Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
7
|
Xue L, Chen R, Liu Y, Niu P, Zhou J, Liu J, Zhang J, Chen H. Association of maternal blood high-mobility group box 1 levels and adverse pregnancy outcomes: A systematic review and meta-analysis. Reprod Biol 2024; 24:100859. [PMID: 38492434 DOI: 10.1016/j.repbio.2024.100859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 03/18/2024]
Abstract
Conflicting findings have emerged regarding the levels of high mobility group box 1 (HMGB1) in individuals experiencing adverse pregnancy outcomes. Here we conducted a meta-analysis to assess the association between maternal blood HMGB1 levels and adverse pregnancy outcomes. Utilizing databases such as PubMed, Cochrane Central Register of Controlled Trials, Web of Science, Embase and China National Knowledge Infrastructure (CNKI), a systematic literature search was conducted in January 2024. Eligible literature was screened according to inclusion and exclusion criteria. Quality assessment was evaluated using the Newcastle-Ottawa Scale (NOS). The extracted data were analyzed using Review Manager 5.4 and STATA 12.0 software. 21 observational studies with a total of 2471 participants were included in this meta-analysis. Significantly higher peripheral blood levels of HMGB1 were associated with preeclampsia (PE) (SMD=1.34; 95% CI: 0.72-1.95; P < 0.0001) and gestational diabetes mellitus (GDM) (SMD=1.20; 95% CI: 0.31-2.09; P = 0.009). Additionally, HMGB1 levels in peripheral blood were significantly elevated in patients with unexplained recurrent spontaneous abortion (URSA) than those in pregnancy controls (SMD=4.22; 95% CI: 1.64-6.80; P = 0.001) or non-pregnancy controls (SMD=3.87; 95% CI: 1.81-5.92; P = 0.0002). Interestingly, higher blood HMGB1 levels were observed in women with preterm birth (PTB), however, the results did not reach a statistical difference (SMD=0.54; 95% CI: -0.36-1.44; P = 0.24). In conclusion, overexpressed maternal blood HMGB1 levels were associated with adverse pregnancy outcomes, including PE, GDM and URSA. Further studies should be conducted to validate the efficacy of HMGB1 as a biomarker for assessing the risk of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Liping Xue
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Ruolin Chen
- College of Letter and Science, University of California Davis, CA, USA
| | - Ying Liu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Peiguang Niu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Jintuo Zhou
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Jinhua Liu
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China
| | - Jinhua Zhang
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| | - Huajiao Chen
- Department of Pharmacy, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China; School of Pharmacy, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
8
|
Murthi P, Harris LK. Liposome-Encapsulated Anti-inflammatory Proteins for Targeted Delivery to the Placenta to Treat Fetal Growth Restriction. Methods Mol Biol 2024; 2728:165-172. [PMID: 38019400 DOI: 10.1007/978-1-0716-3495-0_14] [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] [Indexed: 11/30/2023]
Abstract
Fetal growth restriction (FGR), the failure of a fetus to reach its genetically determined growth potential, is a serious complication affecting up to 10% of pregnancies. FGR is a major risk factor for stillbirth and, in the survivors, neurodevelopmental disorders. We have recently identified that the anti-inflammatory and pro-resolving molecule, lipoxin A4 (LXA4) and its soluble receptor, formyl-peptide receptor-2 (FPR-2) are significantly decreased in human placentas from FGR pregnancy. The LXA4 synthetic analog Compound 43 (C43) is considered a safe, anti-inflammatory therapy and is being developed as a treatment for disease conditions with an inflammatory basis, for example, asthma in children. Identification of therapies to treat FGR in utero comes with the need to mitigate their potential side effects and the use of nanoparticle-mediated delivery systems could facilitate this. Our current studies are focused on targeting the resolution of inflammation observed in FGR placentas, by synthesizing liposome-encapsulated C43 as a novel therapeutic to improve placental function in FGR. In this chapter, we provide a detailed methodological procedure for the preparation of liposomes and conjugation of the peptide sequences, which selectively bind to the outer placental syncytiotrophoblast layer or the vascular endothelium of the uterine spiral arterioles.
Collapse
Affiliation(s)
- Padma Murthi
- Department of Pharmacology, Monash University, Clayton, VIC, Australia.
- Department of Obstetrics and Gynaecology, University of Melbourne, Melbourne, VIC, Australia.
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, Royal Women's Hospital, Parkville, VIC, Australia.
| | - Lynda K Harris
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, St Mary's Hospital, Manchester, UK
- St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Wang D, Tian Y, Wang S, Li Y, Li H, Jiang N, Xie Y, Yu M, Li A, Wang X, Zhou Q. Antisense oligonuleotides influences trophoblasts behaviors by changing LncNR_040117 expression in antiphospholipid antibody syndrome-induced recurrent pregnancy loss. J Matern Fetal Neonatal Med 2023; 36:2183083. [PMID: 37080915 DOI: 10.1080/14767058.2023.2183083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
OBJECTIVE The primary objective of this study was to explore whether antisense oligonucleotides (ASOs) that reduce LncNR_040117 expression in patients with antiphospholipid antibody syndrome (APS)-induced recurrent pregnancy loss (RPL), and further decrease apoptosis and improve trophoblasts invasion through mitogen-activated protein kinase (MAPK) pathways. This paper aimed to provide a new strategy to treat APS-induced RPL. METHODS In this study, we used quantitative reverse transcription-polymerase chain reaction (RT-qPCR) to analyze the expression level of LncNR 040117 in HTR-8/SVneo cells following transfection with ASOs. Then we utilized Western blotting to test the expression levels of interleukin-1β (IL-1β), intracellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and key molecules of MAPK pathways, including the extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinases (JNK) and p38. In addition, we examined the HTR-8/SVneo cells apoptosis by cell apoptosis assay, and migration and invasion by transwell antibody assay. Each experiment was repeated three times. The data are presented as the means ± SDs, and statistical comparisons were performed using Student's t-test. p < 0.05 was considered significant. RESULT Transfected with ASOs, LncNR_040117 was downregulated in trophoblasts compared with APS-induced RPL patients. And LncNR_040117 low expression induced IL-1β and downstream adhesion molecules ICAM-1 and VCAM-1expression level decreased, as well as MAPK pathways downregulation, including the ERK pathway, JNK pathway and p38/MAPK pathway. Furthermore, all these changes resulted in decreased apoptosis and increased migration and invasion of trophoblasts. CONCLUSION This study indicated that ASOs that decrease LncNR_040117 expression can reduce apoptosis and enhance the invasion and migration of trophoblasts by regulating the MAPK pathway.
Collapse
Affiliation(s)
- Di Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Yijia Tian
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Shan Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yilei Li
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Hao Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Ning Jiang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yu Xie
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Mengru Yu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Aihua Li
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
| | - Xietong Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
- Department of Clinical Laboratory, The Laboratory of Placenta-related Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health and Family Planning Commission of China, Jinan, Shandong, P.R. China
| | - Qian Zhou
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, P.R. China
- Department of Obstetrics and Gynecology, Liaocheng People's Hospital, Liaocheng, Shandong, P.R. China
- Department of Clinical Laboratory, The Laboratory of Placenta-related Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health and Family Planning Commission of China, Jinan, Shandong, P.R. China
| |
Collapse
|
10
|
Gong GS, Muyayalo KP, Zhang YJ, Lin XX, Liao AH. Flip a coin: cell senescence at the maternal-fetal interface†. Biol Reprod 2023; 109:244-255. [PMID: 37402700 DOI: 10.1093/biolre/ioad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/06/2023] Open
Abstract
During pregnancy, cell senescence at the maternal-fetal interface is required for maternal well-being, placental development, and fetal growth. However, recent reports have shown that aberrant cell senescence is associated with multiple pregnancy-associated abnormalities, such as preeclampsia, fetal growth restrictions, recurrent pregnancy loss, and preterm birth. Therefore, the role and impact of cell senescence during pregnancy requires further comprehension. In this review, we discuss the principal role of cell senescence at the maternal-fetal interface, emphasizing its "bright side" during decidualization, placentation, and parturition. In addition, we highlight the impact of its deregulation and how this "dark side" promotes pregnancy-associated abnormalities. Furthermore, we discuss novel and less invasive therapeutic practices associated with the modulation of cell senescence during pregnancy.
Collapse
Affiliation(s)
- Guang-Shun Gong
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Kahindo P Muyayalo
- Department of Obstetrics and Gynecology, University of Kinshasa, Kinshasa, D.R. Congo
| | - Yu-Jing Zhang
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Xin-Xiu Lin
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | - Ai-Hua Liao
- Institute of Reproductive Health, Center for Reproductive Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| |
Collapse
|
11
|
Stenhouse C, Bazer FW, Ashworth CJ. Sexual dimorphism in placental development and function: Comparative physiology with an emphasis on the pig. Mol Reprod Dev 2023; 90:684-696. [PMID: 35466463 DOI: 10.1002/mrd.23573] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022]
Abstract
Across mammalian species, it has been demonstrated that sex influences birth weight, with males being heavier than females; a characteristic that can be observed from early gestation. Male piglets are more likely to be stillborn and have greater preweaning mortality than their female littermates, despite the additional maternal investment into male fetal growth. Given the conserved nature of the genome between the sexes, it is hypothesized that these developmental differences between males and females are most likely orchestrated by differential placental adaptation. This review summarizes the current understanding of fetal sex-specific differences in placental and endometrial structure and function, with an emphasis on pathways found to be differentially regulated in the pig including angiogenesis, apoptosis, and proliferation. Given the importance of piglet sex in agricultural enterprises, and the potential for skewed litter sex ratios, it is imperative to improve understanding of the relationship between fetal sex and molecular signaling in both the placenta and endometria across gestation.
Collapse
Affiliation(s)
- Claire Stenhouse
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
- Functional Genetics and Development Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas, USA
| | - Cheryl J Ashworth
- Functional Genetics and Development Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| |
Collapse
|
12
|
Kajdy A, Sys D, Modzelewski J, Bogusławska J, Cymbaluk-Płoska A, Kwiatkowska E, Bednarek-Jędrzejek M, Borowski D, Stefańska K, Rabijewski M, Baran A, Torbe A, Feduniw S, Kwiatkowski S. Evidence of Placental Aging in Late SGA, Fetal Growth Restriction and Stillbirth-A Systematic Review. Biomedicines 2023; 11:1785. [PMID: 37509425 PMCID: PMC10376283 DOI: 10.3390/biomedicines11071785] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
During pregnancy, the placenta undergoes a natural aging process, which is considered normal. However, it has been hypothesized that an abnormally accelerated and premature aging of the placenta may contribute to placenta-related health issues. Placental senescence has been linked to several obstetric complications, including abnormal fetal growth, preeclampsia, preterm birth, and stillbirth, with stillbirth being the most challenging. A systematic search was conducted on Pubmed, Embase, and Scopus databases. Twenty-two full-text articles were identified for the final synthesis. Of these, 15 presented original research and 7 presented narrative reviews. There is a paucity of evidence in the literature on the role of placental aging in late small for gestational age (SGA), fetal growth restriction (FGR), and stillbirth. For future research, guidelines for both planning and reporting research must be implemented. The inclusion criteria should include clear differentiation between early and late SGA and FGR. As for stillbirths, only those with no other known cause of stillbirth should be included in the studies. This means excluding stillbirths due to congenital defects, infections, placental abruption, and maternal conditions affecting feto-maternal hemodynamics.
Collapse
Affiliation(s)
- Anna Kajdy
- First Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 01-004 Warsaw, Poland
| | - Dorota Sys
- Department of Medical Statistics, School of Public Health, Centre of Postgraduate Medical Education, 01-828 Warsaw, Poland
| | - Jan Modzelewski
- First Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 01-004 Warsaw, Poland
| | - Joanna Bogusławska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01-813 Warsaw, Poland
| | - Aneta Cymbaluk-Płoska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland
| | | | - Dariusz Borowski
- Clinic of Obstetrics and Gynaecology, Provincial Combined Hospital in Kielce, 25-736 Kielce, Poland
| | - Katarzyna Stefańska
- Department of Obstetrics, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Michał Rabijewski
- Department of Reproductive Health, Center of Postgraduate Medical Education, Centre of Postgraduate Medical Education, Żelazna 90 St., 01-004 Warsaw, Poland
| | - Arkadiusz Baran
- First Department of Obstetrics and Gynecology, Centre of Postgraduate Medical Education, 01-004 Warsaw, Poland
| | - Andrzej Torbe
- Department Obstetrics and Gynecology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Stepan Feduniw
- Department of Gynecology, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Sebastian Kwiatkowski
- Department Obstetrics and Gynecology, Pomeranian Medical University, 70-111 Szczecin, Poland
| |
Collapse
|
13
|
Wang R, Zhao J, Liu C, Li S, Liu W, Cao Q. Decreased AGGF1 facilitates the progression of placenta accreta spectrum via mediating the P53 signaling pathway under the regulation of miR-1296-5p. Reprod Biol 2023; 23:100735. [PMID: 36753931 DOI: 10.1016/j.repbio.2023.100735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 02/09/2023]
Abstract
Placenta accreta spectrum (PAS), an emerging health issue worldwide, is the major causative factor of maternal morbidity and mortality in modern obstetrics, but limited studies have contributed to our understanding of the molecular biology of PAS. This study addressed the expression of AGGF1 and its specific role in the etiology of PAS. The expression of AGGF1 in the placentas of PAS was determined by quantitative PCR, western blot and immunohistochemistry. CCK-8 assay, wound healing assay, Transwell invasion assay and flow cytometry assay were performed to monitor cell proliferation, migration, invasion and apoptosis. The interaction between miR-1296-5p and AGGF1 was detected by dual-luciferase reporter gene assay. Results showed that the mRNA and protein expression of AGGF1 was decremented in placental tissues of PAS patients, compared with samples from women with placenta previa and normal pregnant women. Downregulation of AGGF1 promoted cell proliferation, invasion and migration, inhibited apoptosis in vitro, decreased P53 and Bax expression, and simultaneously increased Bcl-2 expression, whereas overexpression of AGGF1 had the opposite results. Additionally, the dual-luciferase assay confirmed AGGF1 as a target gene of miR-1296-5p in placental tissues of PAS. Particularly, miR-1296-5p fostered HTR8/SVneo cell proliferation, invasion, repression of apoptosis and regulation of P53 signaling axis by downregulating AGGF1 expression. Collectively, our study accentuated that downregulation of placental AGGF1 promoted trophoblast over-invasion by mediating the P53 signaling pathway under the regulation of miR-1296-5p.
Collapse
Affiliation(s)
- Runfang Wang
- Department of Obstetrics and Gynecology, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jing Zhao
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Cuilian Liu
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Shengxian Li
- Department of Obstetrics and Gynecology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Weifang Liu
- Department of Obstetrics and Gynecology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Qinying Cao
- Department of Obstetrics and Gynecology, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Obstetrics and Gynecology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China.
| |
Collapse
|
14
|
Scarpato R, Colosimo V, Chiaramonte A, Di Bello D, Esposti V, Falaschi A, Ghirri P, Micheli C, Testi S. High level of γH2AX phosphorylation in the cord-blood cells of large-for-gestational-age (LGA) newborns. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2022; 881:503526. [PMID: 36031337 DOI: 10.1016/j.mrgentox.2022.503526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 06/15/2023]
Abstract
Newborns can experience adverse effects as a consequence of maternal or in utero exposure, altered growth of the fetus, or placental dysfunctions. Accurate characterization of gestational age allows monitoring of fetal growth, identification of deviations from the normal growth trajectory, and classification of babies as adapted, small, or large for gestational age (AGA, SGA, or LGA). The aim of this work was to evaluate nuclear and oxidative damage in umbilical cord-blood cells of newborns (sampled at birth), by applying the γH2AX assay and the fluorescent probe BODIPY581/591 C11, to detect DNA DSB and cell membrane oxidation, respectively. No statistically significant differences were observed in the proportion of oxidized cord-blood cells among the groups of newborns, although the LGA group showed the highest value. With regard to genome damage, elevated levels of γH2AX foci were detected in the cell nuclei from LGA newborns as compared to AGA or SGA babies, whose values did not differ from each other. Considering that the observed DNA damage, although still repairable, can represent a risk factor for obesity, metabolic diseases, or other pathologies, monitoring genome and cell integrity at birth can provide useful information for prevention of diseases later in life.
Collapse
Affiliation(s)
- Roberto Scarpato
- Unità di Genetica, Dipartimento di Biologia, University of Pisa, Pisa, Italy.
| | - Valentina Colosimo
- Unità di Genetica, Dipartimento di Biologia, University of Pisa, Pisa, Italy
| | - Anna Chiaramonte
- Unità di Genetica, Dipartimento di Biologia, University of Pisa, Pisa, Italy; Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Women-Child-Newborn Obstetrics and Gynaecology, Milano, Italy
| | - Domenica Di Bello
- Unità di Genetica, Dipartimento di Biologia, University of Pisa, Pisa, Italy
| | - Veronica Esposti
- Unità di Genetica, Dipartimento di Biologia, University of Pisa, Pisa, Italy
| | - Aurora Falaschi
- Unità di Genetica, Dipartimento di Biologia, University of Pisa, Pisa, Italy
| | - Paolo Ghirri
- Division of Neonatology and NICU, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Consuelo Micheli
- Unità di Genetica, Dipartimento di Biologia, University of Pisa, Pisa, Italy
| | - Serena Testi
- Unità di Genetica, Dipartimento di Biologia, University of Pisa, Pisa, Italy
| |
Collapse
|
15
|
Zou Z, Harris LK, Forbes K, Heazell AEP. Placental expression of Estrogen related receptor gamma (ESRRG) is reduced in FGR pregnancies and is mediated by hypoxia. Biol Reprod 2022; 107:846-857. [PMID: 35594451 PMCID: PMC9476228 DOI: 10.1093/biolre/ioac108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/12/2022] [Accepted: 05/13/2021] [Indexed: 11/16/2022] Open
Abstract
Fetal growth restriction (FGR) describes a fetus which has not achieved its genetic growth potential; it is closely linked to placental dysfunction and uteroplacental hypoxia. Estrogen-related receptor gamma (ESRRG) is regulated by hypoxia and is highly expressed in the placenta. We hypothesized ESRRG is a regulator of hypoxia-mediated placental dysfunction in FGR pregnancies. Placentas were collected from women delivering appropriate for gestational age (AGA; n = 14) or FGR (n = 14) infants. Placental explants (n = 15) from uncomplicated pregnancies were cultured for up to 4 days in 21% or 1% O2, or with 200 μM cobalt chloride (CoCl2), or treated with the ESRRG agonists DY131 under different oxygen concentrations. RT-PCR, Western blotting, and immunochemistry were used to assess mRNA and protein levels of ESRRG and its localization in placental tissue from FGR or AGA pregnancies, and in cultured placental explants. ESRRG mRNA and protein expression were significantly reduced in FGR placentas, as was mRNA expression of the downstream targets of ESRRG, hydroxysteroid 11-beta dehydrogenase 2 (HSD11B2), and cytochrome P-450 (CYP19A1.1). Hypoxia-inducible factor 1-alpha protein localized to the nuclei of the cytotrophoblasts and stromal cells in the explants exposed to CoCl2 or 1% O2. Both hypoxia and CoCl2 treatment decreased ESRRG and its downstream genes’ mRNA expression, but not ESRRG protein expression. DY131 increased the expression of ESRRG signaling pathways and prevented abnormal cell turnover induced by hypoxia. These data show that placental ESRRG is hypoxia-sensitive and altered ESRRG-mediated signaling may contribute to hypoxia-induced placental dysfunction in FGR. Furthermore, DY131 could be used as a novel therapeutic approach for the treatment of placental dysfunction.
Collapse
Affiliation(s)
- Zhiyong Zou
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 5th floor (Research), St Mary's Hospital, Oxford Road, Manchester, UK, M13 9WL.,St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UK
| | - Lynda K Harris
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 5th floor (Research), St Mary's Hospital, Oxford Road, Manchester, UK, M13 9WL.,St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UK.,Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Karen Forbes
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 5th floor (Research), St Mary's Hospital, Oxford Road, Manchester, UK, M13 9WL.,St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UK.,Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - Alexander E P Heazell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, Faculty of Biology, Medicine and Health, University of Manchester, 5th floor (Research), St Mary's Hospital, Oxford Road, Manchester, UK, M13 9WL.,St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UK
| |
Collapse
|
16
|
Zou Z, Harris LK, Forbes K, Heazell AEP. Sex-specific effects of Bisphenol a on the signalling pathway of ESRRG in the human placenta. Biol Reprod 2022; 106:1278-1291. [PMID: 35220427 PMCID: PMC9198953 DOI: 10.1093/biolre/ioac044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 01/17/2022] [Accepted: 02/16/2022] [Indexed: 11/17/2022] Open
Abstract
Bisphenol A (BPA) exposure during pregnancy is associated with low fetal weight, particularly in male fetuses. The expression of estrogen-related receptor gamma (ESRRG), a receptor for BPA in the human placenta, is reduced in fetal growth restriction. This study sought to explore whether ESRRG signaling mediates BPA-induced placental dysfunction and determine whether changes in the ESRRG signaling pathway are sex-specific. Placental villous explants from 18 normal term pregnancies were cultured with a range of BPA concentrations (1 nM–1 μM). Baseline BPA concentrations in the placental tissue used for explant culture ranged from 0.04 to 5.1 nM (average 2.3 ±1.9 nM; n = 6). Expression of ESRRG signaling pathway constituents and cell turnover were quantified. BPA (1 μM) increased ESRRG mRNA expression after 24 h in both sexes. ESRRG mRNA and protein expression was increased in female placentas treated with 1 μM BPA for 24 h but was decreased in male placentas treated with 1 nM or 1 μM for 48 h. Levels of 17β-hydroxysteroid dehydrogenase type 1 (HSD17B1) and placenta specific-1 (PLAC1), genes downstream of ESRRG, were also affected. HSD17B1 mRNA expression was increased in female placentas by 1 μM BPA; however, 1 nM BPA reduced HSD17B1 and PLAC1 expression in male placentas at 48 h. BPA treatment did not affect rates of proliferation, apoptosis, or syncytiotrophoblast differentiation in cultured villous explants. This study has demonstrated that BPA affects the ESRRG signaling pathway in a sex-specific manner in human placentas and a possible biological mechanism to explain the differential effects of BPA exposure on male and female fetuses observed in epidemiological studies.
Collapse
Affiliation(s)
- Zhiyong Zou
- Maternal and Fetal Health Research Centre, University of Manchester, 5th floor (Research), St Mary's Hospital, Oxford Road, Manchester, UK, M13 9WL
| | - Lynda K Harris
- Maternal and Fetal Health Research Centre, University of Manchester, 5th floor (Research), St Mary's Hospital, Oxford Road, Manchester, UK, M13 9WL
- Division of Pharmacy and Optometry, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Karen Forbes
- Maternal and Fetal Health Research Centre, University of Manchester, 5th floor (Research), St Mary's Hospital, Oxford Road, Manchester, UK, M13 9WL
- Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - Alexander E P Heazell
- Maternal and Fetal Health Research Centre, University of Manchester, 5th floor (Research), St Mary's Hospital, Oxford Road, Manchester, UK, M13 9WL
- St Mary’s Hospital, Manchester Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
17
|
Redman CW, Staff AC, Roberts JM. Syncytiotrophoblast stress in preeclampsia: the convergence point for multiple pathways. Am J Obstet Gynecol 2022; 226:S907-S927. [PMID: 33546842 DOI: 10.1016/j.ajog.2020.09.047] [Citation(s) in RCA: 186] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/14/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022]
Abstract
Preeclampsia evolves in 2 stages: a placental problem that generates signals to the mother to cause a range of responses that comprise the second stage (preeclampsia syndrome). The first stage of early-onset preeclampsia is poor placentation, which we here call malplacentation. The spiral arteries are incompletely remodeled, leading to later placental malperfusion, relatively early in the second half of pregnancy. The long duration of the first stage (several months) is unsurprisingly associated with fetal growth restriction. The first stage of late-onset preeclampsia, approximately 80% of total cases, is shorter (several weeks) and part of a process that is common to all pregnancies. Placental function declines as it outgrows uterine capacity, with increasing chorionic villous packing, compression of the intervillous space, and fetal hypoxia, and causes late-onset clinical presentations such as "unexplained" stillbirths, late-onset fetal growth restriction, or preeclampsia. The second stages of early- and late-onset preeclampsia share syncytiotrophoblast stress as the most relevant feature that causes the maternal syndrome. Syncytiotrophoblast stress signals in the maternal circulation are probably the most specific biomarkers for preeclampsia. In addition, soluble fms-like tyrosine kinase-1 (mainly produced by syncytiotrophoblast) is the best-known biomarker and is routinely used in clinical practice in many locations. How the stress signals change over time in normal pregnancies indicates that syncytiotrophoblast stress begins on average at 30 to 32 weeks' gestation and progresses to term. At term, syncytiotrophoblast shows increasing markers of stress, including apoptosis, pyroptosis, autophagy, syncytial knots, and necrosis. We label this phenotype the "twilight placenta" and argue that it accounts for the clinical problems of postmature pregnancies. Senescence as a stress response differs in multinuclear syncytiotrophoblast from that of mononuclear cells. Syncytiotrophoblast irreversibly acquires part of the senescence phenotype (cell cycle arrest) when it is formed by cell fusion. The 2 pathways converge on the common pathologic endpoint, syncytiotrophoblast stress, and contribute to preeclampsia subtypes. We highlight that the well-known heterogeneity of the preeclampsia syndrome arises from different pathways to this common endpoint, influenced by maternal genetics, epigenetics, lifestyle, and environmental factors with different fetal and maternal responses to the ensuing insults. This complexity mandates a reassessment of our approach to predicting and preventing preeclampsia, and we summarize research priorities to maximize what we can learn about these important issues.
Collapse
|
18
|
Baker BC, Lui S, Lorne I, Heazell AEP, Forbes K, Jones RL. Sexually dimorphic patterns in maternal circulating microRNAs in pregnancies complicated by fetal growth restriction. Biol Sex Differ 2021; 12:61. [PMID: 34789323 PMCID: PMC8597318 DOI: 10.1186/s13293-021-00405-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/27/2021] [Indexed: 12/18/2022] Open
Abstract
Background Current methods fail to accurately predict women at greatest risk of developing fetal growth restriction (FGR) or related adverse outcomes, including stillbirth. Sexual dimorphism in these adverse pregnancy outcomes is well documented as are sex-specific differences in gene and protein expression in the placenta. Circulating maternal serum microRNAs (miRNAs) offer potential as biomarkers that may also be informative of underlying pathology. We hypothesised that FGR would be associated with an altered miRNA profile and would differ depending on fetal sex. Methods miRNA expression profiles were assessed in maternal serum (> 36 weeks’ gestation) from women delivering a severely FGR infant (defined as an individualised birthweight centile (IBC) < 3rd) and matched control participants (AGA; IBC = 20–80th), using miRNA arrays. qPCR was performed using specific miRNA primers in an expanded cohort of patients with IBC < 5th (n = 15 males, n = 16 females/group). Maternal serum human placental lactogen (hPL) was used as a proxy to determine if serum miRNAs were related to placental dysfunction. In silico analyses were performed to predict the potential functions of altered miRNAs. Results Initial analyses revealed 11 miRNAs were altered in maternal serum from FGR pregnancies. In silico analyses revealed all 11 altered miRNAs were located in a network of genes that regulate placental function. Subsequent analysis demonstrated four miRNAs showed sexually dimorphic patterns. miR-28-5p was reduced in FGR pregnancies (p < 0.01) only when there was a female offspring and miR-301a-3p was only reduced in FGR pregnancies with a male fetus (p < 0.05). miR-454-3p was decreased in FGR pregnancies (p < 0.05) regardless of fetal sex but was only positively correlated to hPL when the fetus was female. Conversely, miR-29c-3p was correlated to maternal hPL only when the fetus was male. Target genes for sexually dimorphic miRNAs reveal potential functional roles in the placenta including angiogenesis, placental growth, nutrient transport and apoptosis. Conclusions These studies have identified sexually dimorphic patterns for miRNAs in maternal serum in FGR. These miRNAs may have potential as non-invasive biomarkers for FGR and associated placental dysfunction. Further studies to determine if these miRNAs have potential functional roles in the placenta may provide greater understanding of the pathogenesis of placental dysfunction and the differing susceptibility of male and female fetuses to adverse in utero conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s13293-021-00405-z. Detection and treatment of pregnancies at high risk of fetal growth restriction (FGR) and stillbirth remains a major obstetric challenge; circulating maternal serum microRNAs (miRNAs) offer potential as novel biomarkers. Unbiased analysis of serum miRNAs in women in late pregnancy identified a specific profile of circulating miRNAs in women with a growth-restricted infant. Some altered miRNAs (miR-28-5p, miR-301a-3p) showed sexually dimorphic expression in FGR pregnancies and others a fetal-sex dependent association to a hormonal marker of placental dysfunction (miR-454-3p, miR-29c-3p). miR-301a-3p and miR-28-5p could potentially be used to predict FGR specifically in pregnancies with a male or female baby, respectively, however larger cohort studies are required. Further investigations of these miRNAs and their relationship to placental dysfunction will lead to a better understanding of the pathophysiology of FGR and why there is differing susceptibility of male and female fetuses to FGR and stillbirth.
Collapse
Affiliation(s)
- Bernadette C Baker
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK.
| | - Sylvia Lui
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK.,Division of Inflammation and Repair, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Isabel Lorne
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | - Alexander E P Heazell
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK.,St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UK
| | - Karen Forbes
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK.
| | - Rebecca L Jones
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
19
|
Targeted Delivery of Epidermal Growth Factor to the Human Placenta to Treat Fetal Growth Restriction. Pharmaceutics 2021; 13:pharmaceutics13111778. [PMID: 34834193 PMCID: PMC8618188 DOI: 10.3390/pharmaceutics13111778] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 12/21/2022] Open
Abstract
Placental dysfunction is the underlying cause of pregnancy complications such as fetal growth restriction (FGR) and pre-eclampsia. No therapies are available to treat a poorly functioning placenta, primarily due to the risks of adverse side effects in both the mother and the fetus resulting from systemic drug delivery. The use of targeted liposomes to selectively deliver payloads to the placenta has the potential to overcome these issues. In this study, we assessed the safety and efficacy of epidermal growth factor (EGF)-loaded, peptide-decorated liposomes to improve different aspects of placental function, using tissue from healthy control pregnancies at term, and pregnancies complicated by FGR. Phage screening identified a peptide sequence, CGPSARAPC (GPS), which selectively homed to mouse placentas in vivo, and bound to the outer syncytiotrophoblast layer of human placental explants ex vivo. GPS-decorated liposomes were prepared containing PBS or EGF (50–100 ng/mL), and placental explants were cultured with liposomes for up to 48 h. Undecorated and GPS-decorated liposomes containing PBS did not affect the basal rate of amino acid transport, human chorionic gonadotropin (hCG) release or cell turnover in placental explants from healthy controls. GPS-decorated liposomes containing EGF significantly increased amino acid transporter activity in healthy control explants, but not in placental explants from women with FGR. hCG secretion and cell turnover were unaffected by EGF delivery; however, differential activation of downstream protein kinases was observed when EGF was delivered via GPS-decorated vs. undecorated liposomes. These data indicate that targeted liposomes represent a safe and useful tool for the development of new therapies for placental dysfunction, recapitulating the effects of free EGF.
Collapse
|
20
|
Kajdy A, Modzelewski J, Cymbaluk-Płoska A, Kwiatkowska E, Bednarek-Jędrzejek M, Borowski D, Stefańska K, Rabijewski M, Torbé A, Kwiatkowski S. Molecular Pathways of Cellular Senescence and Placental Aging in Late Fetal Growth Restriction and Stillbirth. Int J Mol Sci 2021; 22:4186. [PMID: 33919502 PMCID: PMC8072706 DOI: 10.3390/ijms22084186] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/20/2022] Open
Abstract
Abnormally accelerated, premature placental senescence plays a crucial role in the genesis of pregnancy pathologies. Abnormal growth in the third trimester can present as small for gestational age fetuses or fetal growth restriction. One differs from the other by the presence of signs of placental insufficiency and the risk of stillbirth. The majority of stillbirths occur in normally grown fetuses and are classified as "unexplained", which often leads to conclusions that they were unpreventable. The main characteristic of aging is a gradual decline in the function of cells, tissues, and organs. These changes result in the accumulation of senescent cells in mitotic tissues. These cells begin the aging process that disrupts tissues' normal functions by affecting neighboring cells, degrading the extracellular matrix, and reducing tissues' regeneration capacity. Different degrees of abnormal placentation result in the severity of fetal growth restriction and its sequelae, including fetal death. This review aims to present the current knowledge and identify future research directions to understand better placental aging in late fetal growth restriction and unexplained stillbirth. We hypothesized that the final diagnosis of placental insufficiency can be made only using markers of placental senescence.
Collapse
Affiliation(s)
- Anna Kajdy
- Department of Reproductive Health, Centre of Postgraduate Medical Education, Żelazna 90 St., 01-004 Warsaw, Poland; (J.M.); (M.R.)
| | - Jan Modzelewski
- Department of Reproductive Health, Centre of Postgraduate Medical Education, Żelazna 90 St., 01-004 Warsaw, Poland; (J.M.); (M.R.)
| | - Aneta Cymbaluk-Płoska
- Department of Gynecological Surgery and Gynecological Oncology of Adults and Adolescents, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland;
| | - Magdalena Bednarek-Jędrzejek
- Department Obstetrics and Gynecology, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.-J.); (A.T.)
| | - Dariusz Borowski
- Clinic of Fetal-Maternal, Gynecology and Neonatology, Collegium Medicum, Nicolaus Copernicus University in Bydgoszcz, Łukasiewicza 1 St., 85-821 Bydgoszcz, Poland;
| | - Katarzyna Stefańska
- Department of Obstetrics, Medical University of Gdańsk, Mariana Smoluchowskiego 17 St., 80-214 Gdańsk, Poland;
| | - Michał Rabijewski
- Department of Reproductive Health, Centre of Postgraduate Medical Education, Żelazna 90 St., 01-004 Warsaw, Poland; (J.M.); (M.R.)
| | - Andrzej Torbé
- Department Obstetrics and Gynecology, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.-J.); (A.T.)
| | - Sebastian Kwiatkowski
- Department Obstetrics and Gynecology, Pomeranian Medical University, Al. Powstańców Wlkp. 72, 70-111 Szczecin, Poland; (M.B.-J.); (A.T.)
| |
Collapse
|
21
|
Kovács P, Joó JG, Tamás V, Molnár Z, Burik-Hajas D, Bódis J, Kornya L. The role of apoptosis in the complex pathogenesis of the most common obstetrics and gynaecology diseases. Physiol Int 2021; 107:106-119. [PMID: 32491289 DOI: 10.1556/2060.2020.00014] [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: 09/09/2019] [Accepted: 01/07/2020] [Indexed: 11/19/2022]
Abstract
Purpose We aimed to assess the etiological role of apoptotic genes Bcl-2 and Bax in the background of major obstetric and gynaecological diseases. Methods Placental tissue samples were collected from 101 pregnancies with intrauterine growth restriction and 104 pregnancies with premature birth with 140 controll samples from term, eutrophic newborns. In addition, gene expression assessment of the genes Bax and Bcl-2 was performed in 101 uterine leiomyoma tissue samples at our disposal with 110 control cases. Gene expression levels were assessed by PCR method. Results The expression of the Bcl-2 gene was decreased in placental samples with intrauterine growth restriction. Significant overexpression of the proapoptotic Bax gene was detected in samples from premature infants. Antiapoptotic Bcl-2 gene expression was found to be significantly increased in fibroid tissues. Conclusion Apoptosis plays a crucial role in the development of the most common OB/GYN conditions. Decrease in the placental expression of the antiapoptotic gene Bcl-2 may upset the balance of programmed cell death.
Collapse
Affiliation(s)
- P Kovács
- 1Clinical Research Units Hungary, Miskolc, Hungary.,2Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary
| | - József Gábor Joó
- 2Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary.,3First Department of Obstetrics and Gynaecology, Semmelweis University, Budapest, Hungary
| | - V Tamás
- 2Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary
| | - Z Molnár
- 2Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary
| | - D Burik-Hajas
- 2Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary
| | - J Bódis
- 2Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary.,4Hungarian Academy of Sciences, University of Pécs (MTA-PTE), Human Reproduction Scientific Research Group, University of Pécs, Pécs, Hungary
| | - L Kornya
- 2Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary.,5Central Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| |
Collapse
|
22
|
Smirnova A, Mentor A, Ranefall P, Bornehag CG, Brunström B, Mattsson A, Jönsson M. Increased apoptosis, reduced Wnt/β-catenin signaling, and altered tail development in zebrafish embryos exposed to a human-relevant chemical mixture. CHEMOSPHERE 2021; 264:128467. [PMID: 33032226 DOI: 10.1016/j.chemosphere.2020.128467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 06/11/2023]
Abstract
A wide variety of anthropogenic chemicals is detected in humans and wildlife and the health effects of various chemical exposures are not well understood. Early life stages are generally the most susceptible to chemical disruption and developmental exposure can cause disease in adulthood, but the mechanistic understanding of such effects is poor. Within the EU project EDC-MixRisk, a chemical mixture (Mixture G) was identified in the Swedish pregnancy cohort SELMA by the inverse association between levels in women at around gestational week ten with birth weight of their children. This mixture was composed of mono-ethyl phthalate, mono-butyl phthalate, mono-benzyl phthalate, mono-ethylhexyl phthalate, mono-isononyl phthalate, triclosan, perfluorohexane sulfonate, perfluorooctanoic acid, and perfluorooctane sulfonate. In a series of experimental studies, we characterized effects of Mixture G on early development in zebrafish models. Here, we studied apoptosis and Wnt/β-catenin signaling which are two evolutionarily conserved signaling pathways of crucial importance during development. We determined effects on apoptosis by measuring TUNEL staining, caspase-3 activity, and acridine orange staining in wildtype zebrafish embryos, while Wnt/β-catenin signaling was assayed using a transgenic line expressing an EGFP reporter at β-catenin-regulated promoters. We found that Mixture G increased apoptosis, suppressed Wnt/β-catenin signaling in the caudal fin, and altered the shape of the caudal fin at water concentrations only 20-100 times higher than the geometric mean serum concentration in the human cohort. These findings call for awareness that pollutant mixtures like mixture G may interfere with a variety of developmental processes, possibly resulting in adverse health effects.
Collapse
Affiliation(s)
- Anna Smirnova
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden; The Centre for Reproductive Biology in Uppsala (CRU), Uppsala, Sweden
| | - Anna Mentor
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden; The Centre for Reproductive Biology in Uppsala (CRU), Uppsala, Sweden
| | - Petter Ranefall
- SciLifeLab BioImage Informatics Facility, and Dept of Information Technology, Uppsala University, Uppsala, Sweden
| | - Carl-Gustaf Bornehag
- Public Health Sciences, Karlstad University, Karlstad, Sweden; Icahn School of Medicine at Mount Sinai, New York, USA
| | - Björn Brunström
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden; The Centre for Reproductive Biology in Uppsala (CRU), Uppsala, Sweden
| | - Anna Mattsson
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden; The Centre for Reproductive Biology in Uppsala (CRU), Uppsala, Sweden
| | - Maria Jönsson
- Department of Environmental Toxicology, Uppsala University, Uppsala, Sweden; The Centre for Reproductive Biology in Uppsala (CRU), Uppsala, Sweden.
| |
Collapse
|
23
|
Harris LK, Pantham P, Yong HEJ, Pratt A, Borg AJ, Crocker I, Westwood M, Aplin J, Kalionis B, Murthi P. The role of insulin-like growth factor 2 receptor-mediated homeobox gene expression in human placental apoptosis, and its implications in idiopathic fetal growth restriction. Mol Hum Reprod 2020; 25:572-585. [PMID: 31418778 DOI: 10.1093/molehr/gaz047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 07/21/2019] [Indexed: 12/27/2022] Open
Abstract
Fetal growth restriction (FGR) is caused by poor placental development and function early in gestation. It is well known that placentas from women with FGR exhibit reduced cell growth, elevated levels of apoptosis and perturbed expression of the growth factors, cytokines and the homeobox gene family of transcription factors. Previous studies have reported that insulin-like growth factor-2 (IGF2) interacts with its receptor-2 (IGF2R) to regulate villous trophoblast survival and apoptosis. In this study, we hypothesized that human placental IGF2R-mediated homeobox gene expression is altered in FGR and contributes to abnormal trophoblast function. This study was designed to determine the association between IGF2R, homeobox gene expression and cell survival in pregnancies affected by FGR. Third trimester placentas were collected from FGR-affected pregnancies (n = 29) and gestation matched with control pregnancies (n = 30). Functional analyses were then performed in vitro using term placental explants (n = 4) and BeWo trophoblast cells. mRNA expression was determined by real-time PCR, while protein expression was examined by immunoblotting and immunohistochemistry. siRNA transfection was used to silence IGF2R expression in placental explants and the BeWo cell-line. cDNA arrays were used to screen for downstream targets of IGF2R, specifically homeobox gene transcription factors and apoptosis-related genes. Functional effects of silencing IGF2R were then verified by β-hCG ELISA, caspase activity assays and a real-time electrical cell-impedance assay for differentiation, apoptosis and cell growth potential, respectively. IGF2R expression was significantly decreased in placentas from pregnancies complicated by idiopathic FGR (P < 0.05 versus control). siRNA-mediated IGF2R knockdown in term placental explants and the trophoblast cell line BeWo resulted in altered expression of homeobox gene transcription factors, including increased expression of distal-less homeobox gene 5 (DLX5), and decreased expression of H2.0-Like Homeobox 1 (HLX) (P < 0.05 versus control). Knockdown of IGF2R transcription increased the expression and activity of caspase-6 and caspase-8 in placental explants, decreased BeWo proliferation and increased BeWo differentiation (all P < 0.05 compared to respective controls). This is the first study linking IGF2R placental expression with changes in the expression of homeobox genes that control cellular signalling pathways responsible for increased trophoblast cell apoptosis, which is a characteristic feature of FGR.
Collapse
Affiliation(s)
- Lynda K Harris
- Division of Pharmacy and Optometry, The University of Manchester, Stopford Building, Manchester, UK.,Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, St. Mary's Hospital, Manchester, UK
| | - Priyadarshini Pantham
- Department of Obstetrics & Gynaecology, The University of Auckland, Grafton, Auckland, New Zealand.,Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hannah E J Yong
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia.,Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Anita Pratt
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia.,Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Anthony J Borg
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Ian Crocker
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, St. Mary's Hospital, Manchester, UK
| | - Melissa Westwood
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, St. Mary's Hospital, Manchester, UK
| | - John Aplin
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester Academic Health Sciences Centre, St. Mary's Hospital, Manchester, UK
| | - Bill Kalionis
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia.,Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Padma Murthi
- University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia.,Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
24
|
Sharps MC, Baker BC, Guevara T, Bischof H, Jones RL, Greenwood SL, Heazell AEP. Increased placental macrophages and a pro-inflammatory profile in placentas and maternal serum in infants with a decreased growth rate in the third trimester of pregnancy. Am J Reprod Immunol 2020; 84:e13267. [PMID: 32421915 DOI: 10.1111/aji.13267] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/23/2020] [Accepted: 05/13/2020] [Indexed: 12/16/2022] Open
Abstract
PROBLEM There is growing evidence for the role of placental inflammation in the pathophysiology of pregnancy complications including fetal growth restriction (FGR). This study aimed to characterize the inflammatory profile in the maternal circulation and the placenta of infants who were growth restricted and those that were small for gestational age (SGA). METHOD OF STUDY Placental villous tissue and maternal serum were obtained from pregnancies where infants were SGA at birth or who had a decreasing growth rate (≥25 centiles) across the third trimester. Immunohistochemical and histological analyses of placental samples were conducted for macrophage number, alongside vascular and cell turnover analysis. Inflammatory profile was analyzed in maternal and placental compartments via ELISAs and multiplex assays. RESULTS There were significantly more CD163+ macrophages in placentas of infants with a decreased growth rate compared to controls, but not in SGA infants (median 8.6/ nuclei vs 3.8 and 2.9, P = .008 and P = .003, respectively). Uric acid (P = .0007) and IL-8 (P = .0008) were increased in placentas, and S100A8 (P < .0002) was increased in maternal serum of infants with decreased growth rate. No changes in the maternal serum or placental lysates of SGA infants were observed. CONCLUSION The evidence of an altered inflammatory profile in infants with a decreasing growth rate, but not in those that were born SGA, provides further evidence that inflammation plays a role in true FGR. It remains unclear whether the increased placental macrophages occur as a direct result, or as a consequence of the pro-inflammatory environment observed in fetal growth restriction.
Collapse
Affiliation(s)
- Megan C Sharps
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health, Tommy's Maternal and Fetal Health Research Centre, 5th Floor St. Mary's Hospital, University of Manchester, Manchester, UK
| | - Bernadette C Baker
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health, Tommy's Maternal and Fetal Health Research Centre, 5th Floor St. Mary's Hospital, University of Manchester, Manchester, UK
| | - Tatiana Guevara
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health, Tommy's Maternal and Fetal Health Research Centre, 5th Floor St. Mary's Hospital, University of Manchester, Manchester, UK
| | - Helen Bischof
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health, Tommy's Maternal and Fetal Health Research Centre, 5th Floor St. Mary's Hospital, University of Manchester, Manchester, UK
| | - Rebecca L Jones
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health, Tommy's Maternal and Fetal Health Research Centre, 5th Floor St. Mary's Hospital, University of Manchester, Manchester, UK
| | - Susan L Greenwood
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health, Tommy's Maternal and Fetal Health Research Centre, 5th Floor St. Mary's Hospital, University of Manchester, Manchester, UK
| | - Alexander E P Heazell
- Division of Developmental Biology & Medicine, Faculty of Biology, Medicine & Health, Tommy's Maternal and Fetal Health Research Centre, 5th Floor St. Mary's Hospital, University of Manchester, Manchester, UK.,Manchester Academic Health Science Centre, St. Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
25
|
Sun C, Groom KM, Oyston C, Chamley LW, Clark AR, James JL. The placenta in fetal growth restriction: What is going wrong? Placenta 2020; 96:10-18. [PMID: 32421528 DOI: 10.1016/j.placenta.2020.05.003] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/17/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
The placenta is essential for the efficient delivery of nutrients and oxygen from mother to fetus to maintain normal fetal growth. Dysfunctional placental development underpins many pregnancy complications, including fetal growth restriction (FGR) a condition in which the fetus does not reach its growth potential. The FGR placenta is smaller than normal placentae throughout gestation and displays maldevelopment of both the placental villi and the fetal vasculature within these villi. Specialized epithelial cells called trophoblasts exhibit abnormal function and development in FGR placentae. This includes an altered balance between proliferation and apoptotic death, premature cellular senescence, and reduced colonisation of the maternal decidual tissue. Thus, the placenta undergoes aberrant changes at the macroscopic to cellular level in FGR, which can limit exchange capacity and downstream fetal growth. This review aims to compile stereological, in vitro, and imaging data to create a holistic overview of the FGR placenta and its pathophysiology, with a focus on the contribution of trophoblasts.
Collapse
Affiliation(s)
- Cherry Sun
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand.
| | - Katie M Groom
- Liggins Institute, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Charlotte Oyston
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, The University of Auckland, Auckland Bioengineering, House, Level 6/70 Symonds Street, Grafton, Auckland, 1010, New Zealand
| | - Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, 85 Park Road, Grafton, Auckland, 1023, New Zealand
| |
Collapse
|
26
|
Differential expression of several factors involved in placental development in normal and abnormal condition. Placenta 2020; 95:1-8. [PMID: 32339142 DOI: 10.1016/j.placenta.2020.04.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
The placenta, a temporary organ that forms during pregnancy, is the largest fetal organ and the first to develop. It is recognized as an organ that plays a vital role as a metabolic and physical barrier in the fetoplacental unit; throughout fetal development it acts as the lungs, gut, kidneys, and liver of the fetus. When its two components, the fetal and the maternal one, successfully interact, pregnancy proceeds healthily. However, in some cases there may be pregnancy disorders, such as preeclampsia (PE) and gestational diabetes mellitus (GDM), which can lead to a different outcome for the mother and the newborn. In recent years, several studies have been conducted to try to understand how the expression of factors involved in the development of the placenta varies under pathological conditions compared with normal conditions. The purpose of this review is to summarize recent discoveries in this field.
Collapse
|
27
|
Naidoo P, Naidoo RN, Ramkaran P, Chuturgoon AA. Effect of maternal HIV infection, BMI and NOx air pollution exposure on birth outcomes in South African pregnant women genotyped for the p53 Pro72Arg (rs1042522). Int J Immunogenet 2020; 47:414-429. [PMID: 32080966 DOI: 10.1111/iji.12481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/15/2022]
Abstract
Tumour suppressor protein, p53, plays a role in modulating innate immune responses, DNA repair, cell cycle arrest, senescence and apoptosis. Maternal nitrogen oxide (NOx) air pollution exposure, body mass index (BMI), human immunodeficiency virus (HIV) infection and p53 Pro72Arg (rs1042522) affect foetal growth. We investigated whether the aforementioned factors influence birth outcomes in a South African population. Pregnant women (n = 300; HIV -ve = 194 and HIV +ve = 106) were genotyped for the p53 rs1042522 using polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), and further stratified based on HIV status, infants' birthweight (BW; NBW: normal BW [>2,500 g] and LBW: low BW [<2,500 g]) and gestational age (GA; NGA: normal GA [>37 weeks] and PTB: preterm birth [≤37 weeks]). A land use regression model was developed to characterize maternal NOx exposure. Pearson's correlation and multivariate regression analysis statistical tests were used to determine the effect of rs1042522 genotyped pregnant women's BMI and NOx exposure on maternal blood pressure and haemoglobin and iron levels, and infants' anthropometric measurements and Appearance Pulse Grimace Activity and Respiration (APGAR) scores. The prevalence of LBW and PTB was 14.7% and 18.7%, respectively. The LBW group had a higher frequency of the variant Arg-allele versus NBW group (47.7% vs. 31.4%, p = .0046, OR = 2.0, 95% CI = 1.26-3.17). No association was observed between NGA and PTB groups. A significant association between BMI and systolic blood pressure (r = .50, p = .00; B = 0.76, p = .002) and birth length (r = -.28, p = .01; B = -0.107, p = .011), and NOx and birth length (r = -.26, p = .08; B = -0.191, p = .046) and birthweight (B = -8.87, p = .048) was observed in HIV-infected mothers with the variant Pro/Arg + Arg/Arg genotypes. Mothers from the LBW group with the variant genotypes displayed an association between NOx and diastolic blood pressure (r = .58, p = .04), blood iron levels (r = -.60, p = .04; B = -0.204, p = .004), APGAR scores at 1 min (r = -.86, p = .00; B = -0.101, p = .003) and 5 min (r = -.75, p = .01) and birth length (r = -.61, p = .04), and BMI and diastolic blood pressure (r = .72, p = .01). In the PTB group, maternal variant genotypes and NOx were associated with blood haemoglobin levels (B = -0.132, p = .045) and APGAR scores at 1 min (B = -0.161, p = .045) and 5 min (B = -0.147, p = .043). Maternal rs1042522 Arg-allele, HIV infection, BMI and NOx exposure collectively play a role in lowering blood iron levels, gestational hypertension and LBW outcomes.
Collapse
Affiliation(s)
- Pragalathan Naidoo
- Discipline of Medical Biochemistry and Chemical Pathology, Howard College Campus, University of KwaZulu-Natal, Durban, South Africa
| | - Rajen N Naidoo
- Discipline of Occupational and Environmental Health, Howard College Campus, University of KwaZulu-Natal, Durban, South Africa
| | - Prithiksha Ramkaran
- Discipline of Medical Biochemistry and Chemical Pathology, Howard College Campus, University of KwaZulu-Natal, Durban, South Africa
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry and Chemical Pathology, Howard College Campus, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
28
|
Hung TH, Huang SY, Chen SF, Wu CP, Hsieh TT. Decreased placental apoptosis and autophagy in pregnancies complicated by gestational diabetes with large-for-gestational age fetuses. Placenta 2019; 90:27-36. [PMID: 32056548 DOI: 10.1016/j.placenta.2019.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 11/21/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Dysregulation of placental apoptosis and autophagy are observed in pregnancy complications including preeclampsia and fetal growth restriction. However, studies of their changes in the placentas of women with gestational diabetes mellitus (GDM) show inconsistent results. We aimed to compare the changes in apoptosis, autophagy, and Bcl-2 family proteins in the placentas from women with normal pregnancies and those with GDM, with or without large-for-gestational age (LGA) infants and to investigate the effect of hyperglycemia on the changes in apoptosis, autophagy, and Bcl-2 family proteins in primary cytotrophoblastic cells. METHODS Villous tissues were obtained from normal pregnant women and those with GDM, with or without LGA infants. Primary cytotrophoblast cells were isolated from normal term placentas and cultured under standard, hyperglycemic, or hyperosmotic conditions. RESULTS Compared to placentas from normal pregnant women, those from GDM women with LGA infants were heavier, had lower beclin-1 and DRAM levels, less M30 and cleaved PARP immunoreactivity, and increased Ki-67 immunoreactivity. These changes were associated with increased Bcl-xL and decreased Bak levels. Increased glucose concentration led to lower ATG5, beclin-1, LC3B-II, p62, and DRAM levels, lower annexin V and M30-positive cell percentages, and less cleaved PARP changes compared with standard culture conditions. Hyperglycemia caused higher Bcl-xL levels and lower Bak and Bad levels than did standard culture conditions. DISCUSSION There were differential changes in apoptosis and autophagy between placentas from normal pregnant women and those from GDM women with LGA infants. Bcl-2 family proteins are likely involved in the regulation of these changes.
Collapse
Affiliation(s)
- Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan; Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Shih-Yin Huang
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, Department of Physiology and Pharmacology and Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - T'sang-T'ang Hsieh
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
29
|
Doğanlar ZB, Güçlü H, Öztopuz Ö, Türkön H, Dogan A, Uzun M, Doğanlar O. The Role of Melatonin in Oxidative Stress, DNA Damage, Apoptosis and Angiogenesis in Fetal Eye under Preeclampsia and Melatonin Deficiency Stress. Curr Eye Res 2019; 44:1157-1169. [PMID: 31090463 DOI: 10.1080/02713683.2019.1619778] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Aim: The aim of this study was to investigate the possible mechanisms of ocular damage induced by pinealectomy (PNX) and preeclampsia (PE), and to determine the cellular and molecular effects of melatonin treatment on oxidative stress, DNA damage, molecular chaperone responses, induction of apoptosis and angiogenesis in the fetal eye of both PNX and PNX+PE animals. Material and Methods: We analysed therapeutic potential of melatonin on fetal eye damage in PNX and PNX+PE animals using Malondialdehyde (MDA), Random Amplified Polymorphic DNA (RAPD), qRT-PCR and Western blot assays. Results: Our study presents three preliminary findings: (a) in fetal eye tissues, PNX and PNX+PE significantly induce oxidative damage to both DNA and protein contents, leading to a dramatic increase in caspase-dependent apoptotic signalling in both mitochondrial and death receptor pathways; (b) the same conditions trigger hypoxia biomarkers in addition to significant overexpression of HIF1-α, HIF1-β, MMP9 and VEGF genes in the fetal eye; (c) finally, melatonin regulates not only the expression of genes encoding antioxidant enzymes and increase in DNA damage as well as lipid peroxidation but also limits programmed cell death processes in the fetal eye of PNX and PNX+PE animals . Furthermore, melatonin can relatively modulate genes in the HIF1 family, TNF-α and VEGF, thus acting as a direct anti-angiogenic molecule. In conclusion, both PNX and PNX+PE induce ocular damage at both cellular and molecular levels in fetal eye tissue of rats. Conclusion: Our results clearly indicate the potential of melatonin as a preventative therapeutic intervention for fetal ocular damage triggered by both PNX and PNX+PE.
Collapse
Affiliation(s)
- Zeynep Banu Doğanlar
- Department of Medical Biology, Faculty of Medicine, Trakya University , Edirne , Turkey
| | - Hande Güçlü
- Department of Ophthalmology, Faculty of Medicine, Trakya University , Edirne , Turkey
| | - Özlem Öztopuz
- Department of Biophysics, Faculty of Medicine, Çanakkale Onsekiz Mart University , Çanakkale , Turkey
| | - Hakan Türkön
- Department of Biochemistry, Faculty of Medicine, Çanakkale Onsekiz Mart University , Çanakkale , Turkey
| | - Ayten Dogan
- Department of Medical Biology, Faculty of Medicine, Trakya University , Edirne , Turkey
| | - Metehan Uzun
- Department of Physiology, Faculty of Medicine, Çanakkale Onsekiz Mart University , Çanakkale , Turkey
| | - Oguzhan Doğanlar
- Department of Medical Biology, Faculty of Medicine, Trakya University , Edirne , Turkey
| |
Collapse
|
30
|
Lv H, Zhou Q, Li L, Wang S. HLA-C promotes proliferation and cell cycle progression in trophoblast cells. J Matern Fetal Neonatal Med 2019; 34:512-518. [PMID: 31018729 DOI: 10.1080/14767058.2019.1611772] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Objectives: The development of maternal-fetal immune tolerance and adequate trophoblast function are essential for the establishment and maintenance of pregnancy. Human leukocyte antigen (HLA), the major histocompatibility complex (MHC) antigen specific to humans, plays an important role in placentation and is involved in many pregnancy-associated diseases. HLA-C is the only classical MHC I gene expressed at the maternal-fetal interface. To investigate whether HLA-C plays an independent role in regulating the development of trophoblasts, we explored the effect of HLA-C expression on placental development.Methods: The role of HLA-C in the growth and migration of trophoblast JAR and HTR-8/Svneo cell lines was investigated after HLA-C-expressing lentivirus transfection.Results: The MTT assay and colony formation assay showed that HLA-C promoted cell proliferation. Furthermore, cell cycle analysis showed that HLA-C overexpression accelerated the transition of trophoblast cells from the G0/G1 phase to the S phase. However, FACS analysis and migration assay indicated that HLA-C had no significant influence on trophoblast apoptosis and migration.Conclusion: Our study demonstrated for the first time that besides being involved in immune tolerance, HLA-C can directly promote placental growth without interacting with immune cells, which could provide a new insight into studying the functions of HLA-C at the maternal-fetal interface.
Collapse
Affiliation(s)
- Hong Lv
- Center for Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Qian Zhou
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Lie Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shan Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
31
|
Paules C, Dantas AP, Miranda J, Crovetto F, Eixarch E, Rodriguez-Sureda V, Dominguez C, Casu G, Rovira C, Nadal A, Crispi F, Gratacós E. Premature placental aging in term small-for-gestational-age and growth-restricted fetuses. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2019; 53:615-622. [PMID: 30125412 DOI: 10.1002/uog.20103] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/26/2018] [Accepted: 08/10/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE To perform a comprehensive assessment of the placental aging process in small term fetuses classified as being small-for-gestational age (SGA) or having fetal growth restriction (FGR) through analysis of senescence and apoptosis markers. METHODS This was a prospective nested case-control study of singleton pregnancies delivered at term, including 21 control pregnancies with normally grown fetuses and 36 with a small fetus classified as SGA (birth weight between the 3rd and 9th percentiles and normal fetoplacental Doppler; n = 18) or FGR (birth weight < 3rd percentile and/or abnormal cerebroplacental ratio and/or uterine artery Doppler; n = 18). Telomerase activity, telomere length (quantified by comparing the amount of amplification product for the telomere sequence (T) to that of a single copy of the gene 36B4 (S)) and RNA expression of senescence (Sirtuins 1, 3 and 6) and apoptosis (p53, p21, BAX and Caspases 3 and 9) markers (analyzed using the 2-ΔΔCt method) were determined in placental samples collected at birth and compared between the three groups. RESULTS Compared to pregnancies with a normally grown fetus, both SGA and FGR pregnancies presented signs of accelerated placental aging, including lower telomerase activity (mean ± SD, 12.8 ± 6.6% in controls vs 7.98 ± 4.2% in SGA vs 7.79 ± 4.6% in FGR; P = 0.008), shorter telomeres (mean ± SD T/S ratio, 1.20 ± 0.6 in controls vs 1.08 ± 0.9 in SGA vs 0.66 ± 0.5 in FGR; P = 0.047) and reduced Sirtuin-1 RNA expression (mean ± SD 2-ΔΔCt , 1.55 ± 0.8 in controls vs 0.91 ± 0.8 in SGA vs 0.63 ± 0.5 in FGR; P = 0.001) together with increased p53 RNA expression (median (interquartile range) 2-ΔΔCt , 1.07 (0.3-3.3) in controls vs 5.39 (0.6-15) in SGA vs 3.75 (0.9-7.8) in FGR; P = 0.040). FGR cases presented signs of apoptosis, with increased Caspase-3 RNA levels (median (interquartile range) 2-ΔΔCt , 0.94 (0.7-1.7) in controls vs 3.98 (0.9-31) in FGR; P = 0.031) and Caspase-9 RNA levels (median (interquartile range) 2-ΔΔCt , 1.21 (0.6-4.0) in controls vs 3.87 (1.5-9.0) in FGR; P = 0.037) compared with controls. In addition, Sirtuin-1 RNA expression, telomerase activity, telomere length and Caspase-3 activity showed significant linear trends across groups as severity of the condition increased. CONCLUSIONS Accelerated placental aging was observed in both clinical forms of late-onset fetal smallness (SGA and FGR), supporting a common pathophysiology and challenging the concept of SGA fetuses being constitutionally small. Copyright © 2018 ISUOG. Published by John Wiley & Sons Ltd.
Collapse
Affiliation(s)
- C Paules
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, ICGON, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - A P Dantas
- Cardiovascular Institut, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - J Miranda
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, ICGON, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - F Crovetto
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, ICGON, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - E Eixarch
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, ICGON, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Disease (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| | - V Rodriguez-Sureda
- Centre for Biomedical Research on Rare Disease (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
- Biochemistry and Molecular Biology Research Centre for Nanomedicine, Hospital Univeritari Vall d'Hebron, Barcelona, Spain
| | - C Dominguez
- Centre for Biomedical Research on Rare Disease (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
- Biochemistry and Molecular Biology Research Centre for Nanomedicine, Hospital Univeritari Vall d'Hebron, Barcelona, Spain
| | - G Casu
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, ICGON, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - C Rovira
- Department of Pathology, Hospital Sant Joan de Deu, Esplugues de Llobregat, Spain
| | - A Nadal
- Department of Pathology, Hospital Clinic, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - F Crispi
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, ICGON, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Disease (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| | - E Gratacós
- Fetal Medicine Research Center, BCNatal - Barcelona Center for Maternal-Fetal and Neonatal Medicine, Hospital Clínic and Hospital Sant Joan de Deu, ICGON, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
- Centre for Biomedical Research on Rare Disease (CIBER-ER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
32
|
Lappas M, McCracken S, McKelvey K, Lim R, James J, Roberts CT, Fournier T, Alfaidy N, Powell KL, Borg AJ, Morris JM, Leaw B, Singh H, Ebeling PR, Wallace EM, Parry LJ, Dimitriadis E, Murthi P. Formyl peptide receptor-2 is decreased in foetal growth restriction and contributes to placental dysfunction. Mol Hum Reprod 2019; 24:94-109. [PMID: 29272530 DOI: 10.1093/molehr/gax067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Accepted: 12/18/2017] [Indexed: 01/07/2023] Open
Abstract
STUDY QUESTION What is the association between placental formyl peptide receptor 2 (FPR2) and trophoblast and endothelial functions in pregnancies affected by foetal growth restriction (FGR)? SUMMARY ANSWER Reduced FPR2 placental expression in idiopathic FGR results in significantly altered trophoblast differentiation and endothelial function in vitro. WHAT IS KNOWN ALREADY FGR is associated with placental insufficiency, where defective trophoblast and endothelial functions contribute to reduced feto-placental growth. STUDY DESIGN, SIZE, DURATION The expression of FPR2 in placental tissues from human pregnancies complicated with FGR was compared to that in gestation-matched uncomplicated control pregnancies (n = 25 from each group). Fpr2 expression was also determined in placental tissues obtained from a murine model of FGR (n = 4). The functional role of FPR2 in primary trophoblasts and endothelial cells in vitro was assessed in diverse assays in a time-dependent manner. PARTICIPANTS/MATERIALS, SETTING, METHODS Placentae from third-trimester pregnancies complicated by idiopathic FGR (n = 25) and those from gestation-matched pregnancies with appropriately grown infants as controls (n = 25) were collected at gestation 27-40 weeks. Placental tissues were also collected from a spontaneous CBA/CaH × DBA/2 J murine model of FGR. Placental FPR2/Fpr2 mRNA expression was determined by real-time PCR, while protein expression was examined by immunoblotting and immunohistochemistry. siRNA transfection was used to silence FPR2 expression in primary trophoblasts and in human umbilical vein endothelial cells (HUVEC), and the quantitation of cytokines, chemokines and apoptosis was performed following a cDNA array analyses. Functional effects of trophoblast differentiation were measured using HCGB/β-hCG and syncytin-2 expression as well as markers of apoptosis, tumour protein 53 (TP53), caspase 8, B cell lymphoma 2 (BCL2) and BCL associated X (BAX). Endothelial function was assessed by proliferation, network formation and permeability assays. MAIN RESULTS AND THE ROLE OF CHANCE Placental FPR2/Fpr2 expression was significantly decreased in FGR placentae (n = 25, P < 0.05) as well as in murine FGR placentae compared to controls (n = 4, P < 0.05). FPR2 siRNA (siFPR2) in term trophoblasts significantly increased differentiation markers, HCGB and syncytin-2; cytokines, interleukin (IL)-6, CXCL8; and apoptotic markers, TP53, caspase 8 and BAX, but significantly reduced the expression of the chemokines CXCL12 and its receptors CXCR4 and CXCR7; CXCL16 and its receptor, CXCR6; and cytokine, IL-10, compared with control siRNA (siCONT). Treatment of HUVECs with siFPR2 significantly reduced proliferation and endothelial tube formation, but significantly increased permeability of HUVECs. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Reduced expression of placental FPR2/Fpr2 was observed in the third trimester at delivery after development of FGR, suggesting that FPR2 is associated with FGR pregnancies. However, there is a possibility that the decreased placental FPR2 observed in FGR may be a consequence rather than a cause of FGR, although our in vitro functional analyses using primary trophoblasts and endothelial cells suggest that FPR2 may have a direct or indirect regulatory role on trophoblast differentiation and endothelial function in FGR. WIDER IMPLICATIONS OF THE FINDINGS This is the first study linking placental FPR2 expression with changes in the trophoblast and endothelial functions that may explain the placental insufficiency observed in FGR. STUDY FUNDING/COMPETING INTERESTS P.M. and P.R.E. received funding from the Australian Institute of Musculoskeletal Science, Western Health, St. Albans, Victoria 3021, Australia. M.L. is supported by a Career Development Fellowship from the National Health and Medical Research Council (NHMRC; Grant no. 1047025). Monash Health is supported by the Victorian Government's Operational Infrastructure Support Programme. The authors declare that there is no conflict of interest in publishing this work.
Collapse
Affiliation(s)
- Martha Lappas
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria 3079, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Victoria 3079, Australia
| | - Sharon McCracken
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, New South Wales 2065, Australia.,Sydney Medical School Northern, University of Sydney, New South Wales 2006, Australia
| | - Kelly McKelvey
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, New South Wales 2065, Australia.,Sydney Medical School Northern, University of Sydney, New South Wales 2006, Australia
| | - Ratana Lim
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria 3079, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Heidelberg, Victoria 3079, Australia
| | - Joanna James
- Department of Obstetrics and Gynaecology, University of Auckland, New Zealand
| | - Claire T Roberts
- Adelaide Medical School and Robinson Research Institute, University of Adelaide, South Australia 5005, Australia
| | - Thierry Fournier
- INSERM, UMR-S1139, Faculté des Sciences Pharmaceutiques et Biologiques, Paris F-75006, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris F-75006 France.,Fondation PremUp, Paris F-75006, France
| | - Nadia Alfaidy
- Institut National de la Santé et de la Recherche Médicale, Unité 1036, Grenoble, France.,University Grenoble-Alpes, 38000 Grenoble, France.,Commissariat à l'Energie Atomique (CEA), iRTSV- Biology of Cancer and Infection, Grenoble, France
| | - Katie L Powell
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, New South Wales 2065, Australia.,Sydney Medical School Northern, University of Sydney, New South Wales 2006, Australia
| | - Anthony J Borg
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria 3052, Australia
| | - Jonathan M Morris
- Division of Perinatal Research, Kolling Institute, Northern Sydney Local Health District, St Leonards, New South Wales 2065, Australia.,Sydney Medical School Northern, University of Sydney, New South Wales 2006, Australia
| | - Bryan Leaw
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Harmeet Singh
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Peter R Ebeling
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Euan M Wallace
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia
| | - Laura J Parry
- School of Biosciences, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Evdokia Dimitriadis
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Padma Murthi
- Department of Maternal-Fetal Medicine Pregnancy Research Centre, The Royal Women's Hospital, Parkville, Victoria 3052, Australia.,The Ritchie Centre, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.,Department of Medicine, School of Clinical Sciences, Monash University, Clayton, Victoria 3168, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
33
|
Lv Y, Lv M, Ji X, Xue L, Rui C, Yin L, Ding H, Miao Z. Down-regulated expressed protein HMGB3 inhibits proliferation and migration, promotes apoptosis in the placentas of fetal growth restriction. Int J Biochem Cell Biol 2018; 107:69-76. [PMID: 30543931 DOI: 10.1016/j.biocel.2018.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/01/2018] [Accepted: 11/12/2018] [Indexed: 12/26/2022]
Abstract
Fetal growth restriction (FGR) is one of the major complications of pregnancy, which can lead to serious short-term and long-term diseases. High-mobility group box 3 (HMGB3) has been found to contribute to the development of many cancers. However, the role of HMGB3 in the pathogenesis of FGR is blank. Here, we measured the expression level of HMGB3 in the placenta tissues of six normal pregnancies and five FGR patients by western blotting and quantitative real-time polymerase chain reaction (qRT-PCR). CCK8 assay, transwell assay and flow cytometry were used to detect the functional effects of overexpression and silencing of HMGB3 on the HTR8/SVneo trophoblast cell line. The results showed that the protein levels of HMGB3 were significantly decreased in FGR placentas compared to normal controls, while mRNA levels of HMGB3 were not significantly altered. Furthermore, when overexpressed of protein HMGB3 of the trophoblast cells, the proliferation and migration abilities were significantly promoted, and the apoptosis abilities of these cells were statistically inhibited. Cell functional experiments showed the opposite results when the expression of HMGB3 was silent. And the expression of cell function-related genes PCNA, Ki67, Tp53, Bax, MMP-2 and E-cadherin was observed to show corresponding changes by qRT-PCR. The results of mass spectrometry showed that HMGB3 may directly or indirectly interact with 71 proteins. In summary, our results indicated that HMGB3 might be of very great significance to the pathogenesis of FGR and might play the role by leading the dysfunction of placental villous trophoblast cells and through the interaction with some other proteins.
Collapse
Affiliation(s)
- Yan Lv
- Department of Obstetrics and Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Mingming Lv
- Department of Breast, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China; Nanjing Maternal and Child Health Institute, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Xiaohong Ji
- Department of Obstetrics and Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Lu Xue
- Department of Obstetrics and Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Can Rui
- Department of Obstetrics and Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Lingfeng Yin
- Department of Obstetrics and Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China
| | - Hongjuan Ding
- Department of Obstetrics and Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.
| | - Zhijing Miao
- Department of Obstetrics and Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), Nanjing, 210004, China.
| |
Collapse
|
34
|
Pérez-Pérez A, Toro A, Vilariño-Garcia T, Guadix P, Maymó J, Dueñas JL, Varone C, Sánchez-Margalet V. Leptin protects placental cells from apoptosis induced by acidic stress. Cell Tissue Res 2018; 375:733-742. [PMID: 30338379 DOI: 10.1007/s00441-018-2940-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/25/2018] [Indexed: 12/21/2022]
Abstract
Development of the human placenta is critical for a successful pregnancy. The placenta allows the exchange of oxygen and carbon dioxide and is crucial to manage acid-base balance within a narrow pH. It is known that low pH levels are a risk of apoptosis in several tissues. However, there has been little discussion about the effect of acidic stress in the placenta. Leptin is produced by the placenta with a trophic autocrine effect. Previous results of our group have demonstrated that leptin prevents apoptosis of trophoblast cells under different stress conditions such as serum deprivation and hyperthermia. The purpose of the present work is to evaluate acidic stress consequences in trophoblast explant survival and to determine leptin action in these conditions. For this objective, term human trophoblast explants were cultured at physiological pH (pH 7.4) and at acidic pH (pH 6.8) in the presence or absence of leptin. Western blot assays were performed to study the abundance of active caspase-3 and the p89 fragment of PARP-1. Pro-apoptotic and pro-survival members of Bcl-2 family, as Bax, t-Bid, and Bcl-2, were studied. Moreover, p53 pathway was also evaluated including Mdm-2, the main p53 regulator. Active caspase-3 and cleaved PARP-1 abundances were increased at low extracellular pH. Moreover, t-Bid levels were also augmented as well as p53 expression and phosphorylation on S46. Leptin treatment prevents the consequences of acidosis, decreasing p53 expression and increasing Mdm-2 expression. In summary, this work demonstrated for first time that low pH induces apoptosis of human trophoblast explants involving apoptotic intrinsic pathway, and leptin impairs this effect.
Collapse
Affiliation(s)
- Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, Av. Dr. Fedriani 3, 41071, Seville, Spain
| | - Ayelén Toro
- Laboratory of Placental Molecular Physiology, Department of Biological Chemistry, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET, Ciudad Universitaria, Pab. 2, Buenos Aires, Argentina
| | - Teresa Vilariño-Garcia
- Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, Av. Dr. Fedriani 3, 41071, Seville, Spain
| | - Pilar Guadix
- Department of Obstetrics and gynecology, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Julieta Maymó
- Laboratory of Placental Molecular Physiology, Department of Biological Chemistry, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET, Ciudad Universitaria, Pab. 2, Buenos Aires, Argentina
| | - José Luis Dueñas
- Department of Obstetrics and gynecology, Virgen Macarena University Hospital, University of Seville, Seville, Spain
| | - Cecilia Varone
- Laboratory of Placental Molecular Physiology, Department of Biological Chemistry, School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET, Ciudad Universitaria, Pab. 2, Buenos Aires, Argentina
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, Av. Dr. Fedriani 3, 41071, Seville, Spain.
| |
Collapse
|
35
|
Stenhouse C, Hogg CO, Ashworth CJ. Associations between fetal size, sex and both proliferation and apoptosis at the porcine feto-maternal interface. Placenta 2018; 70:15-24. [PMID: 30316322 PMCID: PMC6215148 DOI: 10.1016/j.placenta.2018.08.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 08/17/2018] [Accepted: 08/24/2018] [Indexed: 11/26/2022]
Abstract
Introduction Inadequate fetal growth has severe consequences for both neonatal and adult development. It is hypothesised that the feto-maternal interface associated with the lightest and male fetuses will undergo more apoptosis and less proliferation than those supplying the closest to mean litter weight (CTMLW) and female fetuses respectively. Methods Placental and endometrial samples associated with the lightest and CTMLW (gestational day (GD) 18 and 30), male and female (GD45, 60 and 90) Large White X Landrace conceptuses or fetuses were obtained. The mRNA expression of candidate genes involved in apoptosis or proliferation (BAX, BCL2, P53 and KI67) was quantified by qPCR. TUNEL staining was performed on placental samples supplying the lightest and CTMLW fetuses (GD45 and 60), of both sex (GD60). Results Placentas associated with the lightest fetuses had decreased P53 and KI67 expression compared to the CTMLW fetuses at GD45. At GD60, P53 expression was increased in placentas supplying the lightest compared to CTMLW fetuses. P53 expression was increased in endometrial samples associated with the lightest compared to the CTMLW fetuses at GD45. At GD30 and GD60 respectively, BAX expression was increased and BCL2, P53 and KI67 expression were decreased in endometrial samples associated with females compared to their male littermates. TUNEL staining revealed no association between fetal size or sex, and apoptotic cell number. Discussion This study has highlighted dynamic associations between fetal size, sex, and apoptosis and proliferation at the porcine feto-maternal interface. Further studies should be performed to improve the understanding of the mechanisms behind these findings. Gestational day influence feto-maternal interface apoptotic mRNA expression. Fetal size is associated with feto-maternal interface apoptotic mRNA expression. Sexual dimorphism exists in feto-maternal interface apoptotic mRNA expression.
Collapse
Affiliation(s)
- Claire Stenhouse
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK.
| | - Charis O Hogg
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| | - Cheryl J Ashworth
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, EH25 9RG, UK
| |
Collapse
|
36
|
Gauster M, Maninger S, Siwetz M, Deutsch A, El-Heliebi A, Kolb-Lenz D, Hiden U, Desoye G, Herse F, Prokesch A. Downregulation of p53 drives autophagy during human trophoblast differentiation. Cell Mol Life Sci 2018; 75:1839-1855. [PMID: 29080089 PMCID: PMC5910494 DOI: 10.1007/s00018-017-2695-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/20/2017] [Accepted: 10/24/2017] [Indexed: 01/21/2023]
Abstract
The placental barrier is crucial for the supply of nutrients and oxygen to the developing fetus and is maintained by differentiation and fusion of mononucleated cytotrophoblasts into the syncytiotrophoblast, a process only partially understood. Here transcriptome and pathway analyses during differentiation and fusion of cultured trophoblasts yielded p53 signaling as negative upstream regulator and indicated an upregulation of autophagy-related genes. We further showed p53 mRNA and protein levels decreased during trophoblast differentiation. Reciprocally, autophagic flux increased and cytoplasmic LC3B-GFP puncta became more abundant, indicating enhanced autophagic activity. In line, in human first trimester placenta p53 protein mainly localized to the cytotrophoblast, while autophagy marker LC3B as well as late autophagic compartments were predominantly detectable in the syncytiotrophoblast. Importantly, ectopic overexpression of p53 reduced levels of LC3B-II, supporting a negative regulatory role on autophagy in differentiating trophoblasts. This was also shown in primary trophoblasts and human first trimester placental explants, where pharmacological stabilization of p53 decreased LC3B-II levels. In summary our data suggest that differentiation-dependent downregulation of p53 is a prerequisite for activating autophagy in the syncytiotrophoblast.
Collapse
Affiliation(s)
- Martin Gauster
- Institute of Cell Biology, Histology and Embryology, Medical University Graz, Neue Stiftingtalstraße 6, F/03/38, 8010, Graz, Austria.
| | - Sabine Maninger
- Institute of Cell Biology, Histology and Embryology, Medical University Graz, Neue Stiftingtalstraße 6, F/03/38, 8010, Graz, Austria
| | - Monika Siwetz
- Institute of Cell Biology, Histology and Embryology, Medical University Graz, Neue Stiftingtalstraße 6, F/03/38, 8010, Graz, Austria
| | - Alexander Deutsch
- Division of Hematology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | - Amin El-Heliebi
- Institute of Cell Biology, Histology and Embryology, Medical University Graz, Neue Stiftingtalstraße 6, F/03/38, 8010, Graz, Austria
| | - Dagmar Kolb-Lenz
- Institute of Cell Biology, Histology and Embryology, Medical University Graz, Neue Stiftingtalstraße 6, F/03/38, 8010, Graz, Austria
- Center for Medical Research, Core Facility Ultrastructure Analysis, Medical University Graz, Graz, Austria
| | - Ursula Hiden
- Department of Obstetrics and Gynecology, Medical University Graz, Graz, Austria
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University Graz, Graz, Austria
| | - Florian Herse
- Experimental and Clinical Research Center, A Joint Cooperation Between the Charité Medical Faculty and the Max-Delbrueck Center for Molecular Medicine, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Andreas Prokesch
- Institute of Cell Biology, Histology and Embryology, Medical University Graz, Neue Stiftingtalstraße 6, F/03/38, 8010, Graz, Austria.
| |
Collapse
|
37
|
Dai F, He H, Xu X, Chen S, Wang C, Feng C, Tian Z, Dong H, Xie S. Synthesis and biological evaluation of naphthalimide-polyamine conjugates modified by alkylation as anticancer agents through p53 pathway. Bioorg Chem 2018; 77:16-24. [DOI: 10.1016/j.bioorg.2017.12.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 12/25/2017] [Accepted: 12/30/2017] [Indexed: 12/13/2022]
|
38
|
Muralimanoharan S, Gao X, Weintraub S, Myatt L, Maloyan A. Sexual dimorphism in activation of placental autophagy in obese women with evidence for fetal programming from a placenta-specific mouse model. Autophagy 2018; 12:752-69. [PMID: 26986453 DOI: 10.1080/15548627.2016.1156822] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The incidence of maternal obesity and its co-morbidities (diabetes, cardiovascular disease) continues to increase at an alarming rate, with major public health implications. In utero exposure to maternal obesity has been associated with development of cardiovascular and metabolic diseases in the offspring as a result of developmental programming. The placenta regulates maternal-fetal metabolism and shows significant changes in its function with maternal obesity. Autophagy is a cell-survival process, which is responsible for the degradation of damaged organelles and misfolded proteins. Here we show an activation of autophagosomal formation and autophagosome-lysosome fusion in placentas of males but not females from overweight (OW) and obese (OB) women vs. normal weight (NW) women. However, total autophagic activity in these placentas appeared to be decreased as it showed an increase in SQSTM1/p62 and a decrease in lysosomal biogenesis. A mouse model with a targeted deletion of the essential autophagy gene Atg7 in placental tissue showed significant placental abnormalities comparable to those seen in human placenta with maternal obesity. These included a decrease in expression of mitochondrial genes and antioxidants, and decreased lysosomal biogenesis. Strikingly, the knockout mice were developmentally programmed as they showed an increased sensitivity to high-fat diet-induced obesity, hyperglycemia, hyperinsulinemia, increased adiposity, and cardiac remodeling. In summary, our results indicate a sexual dimorphism in placental autophagy in response to maternal obesity. We also show that autophagy plays an important role in placental function and that inhibition of placental autophagy programs the offspring to obesity, and to metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Sribalasubashini Muralimanoharan
- a Center for Pregnancy and Newborn Research , Department of Obstetrics and Gynecology , University of Texas Health Science Center , San Antonio , TX , USA
| | - Xiaoli Gao
- b The Metabolomics Core Facility, Institutional Mass Spectrometry Laboratory, University of Texas Health Science Center , San Antonio , TX , USA
| | - Susan Weintraub
- b The Metabolomics Core Facility, Institutional Mass Spectrometry Laboratory, University of Texas Health Science Center , San Antonio , TX , USA
| | - Leslie Myatt
- a Center for Pregnancy and Newborn Research , Department of Obstetrics and Gynecology , University of Texas Health Science Center , San Antonio , TX , USA.,c Department of Ob/Gyn , Oregon Health and Science University , Portland , OR , USA
| | - Alina Maloyan
- a Center for Pregnancy and Newborn Research , Department of Obstetrics and Gynecology , University of Texas Health Science Center , San Antonio , TX , USA.,d Knight Cardiovascular Institute, Oregon Health and Science University , Portland , OR , USA
| |
Collapse
|
39
|
Lean SC, Heazell AEP, Dilworth MR, Mills TA, Jones RL. Placental Dysfunction Underlies Increased Risk of Fetal Growth Restriction and Stillbirth in Advanced Maternal Age Women. Sci Rep 2017; 7:9677. [PMID: 28852057 PMCID: PMC5574918 DOI: 10.1038/s41598-017-09814-w] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 07/31/2017] [Indexed: 12/20/2022] Open
Abstract
Pregnancies in women of advanced maternal age (AMA) are susceptible to fetal growth restriction (FGR) and stillbirth. We hypothesised that maternal ageing is associated with utero-placental dysfunction, predisposing to adverse fetal outcomes. Women of AMA (≥35 years) and young controls (20-30 years) with uncomplicated pregnancies were studied. Placentas from AMA women exhibited increased syncytial nuclear aggregates and decreased proliferation, and had increased amino acid transporter activity. Chorionic plate and myometrial artery relaxation was increased compared to controls. AMA was associated with lower maternal serum PAPP-A and sFlt and a higher PlGF:sFlt ratio. AMA mice (38-41 weeks) at E17.5 had fewer pups, more late fetal deaths, reduced fetal weight, increased placental weight and reduced fetal:placental weight ratio compared to 8-12 week controls. Maternofetal clearance of 14C-MeAIB and 3H-taurine was reduced and uterine arteries showed increased relaxation. These studies identify reduced placental efficiency and altered placental function with AMA in women, with evidence of placental adaptations in normal pregnancies. The AMA mouse model complements the human studies, demonstrating high rates of adverse fetal outcomes and commonalities in placental phenotype. These findings highlight placental dysfunction as a potential mechanism for susceptibility to FGR and stillbirth with AMA.
Collapse
Affiliation(s)
- Samantha C Lean
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, University of Manchester, Manchester, United Kingdom.
| | - Alexander E P Heazell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester Academic Health Science Centre, Central Manchester University Hospitals, NHS Foundation Trust, Manchester, United Kingdom
| | - Mark R Dilworth
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester Academic Health Science Centre, Central Manchester University Hospitals, NHS Foundation Trust, Manchester, United Kingdom
| | - Tracey A Mills
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester Academic Health Science Centre, Central Manchester University Hospitals, NHS Foundation Trust, Manchester, United Kingdom
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, University of Manchester, Manchester, United Kingdom
- St. Mary's Hospital, Manchester Academic Health Science Centre, Central Manchester University Hospitals, NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
40
|
Gates KC, Goetzmann LN, Cantlon JD, Jeckel KM, Anthony RV. Effect of proline rich 15-deficiency on trophoblast viability and survival. PLoS One 2017; 12:e0174976. [PMID: 28380025 PMCID: PMC5381842 DOI: 10.1371/journal.pone.0174976] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/17/2017] [Indexed: 01/11/2023] Open
Abstract
Deviations from the normal program of gene expression during early pregnancy can lead to early embryonic loss as well as dysfunctional placentation, which can cause significant morbidity and mortality. Proline rich 15 (PRR15) is a low molecular weight nuclear protein expressed by the trophoblast during early gestation. Lentivirus-mediated knockdown of PRR15 mRNA in ovine trophectoderm led to demise of the embryo by gestational day 15, providing compelling evidence that PRR15 expression is critical during this precarious window of development. Our objective was to determine the effect of PRR15 knockdown on trophoblast gene expression, proliferation, and survival. The first-trimester human trophoblast cell line, ACH-3P, was infected with control lentivirus or a lentivirus expressing a short hairpin (sh)RNA to target PRR15 mRNA for degradation, resulting in a 68% reduction in PRR15 mRNA. Microarray analysis of these cell lines revealed differential expression of genes related to cancer, focal adhesion, and p53 signaling. These changes included significant up-regulation of GDF15, a cytokine increased in pregnancies with preeclampsia. Viability and proliferation decreased in PRR15-deficient cells, which was consistent with down-regulation of cell cycle-related genes CCND1 and CDK6 and an up-regulation of CCNG2 and CDKN1A in the PRR15-deficient cells. TNFSF10, a tumor necrosis factor superfamily member known to induce apoptosis increased significantly in the PRR15-deficient cells. Migration through a basement membrane matrix decreased and an increased population of apoptotic cells was present when treated with shRNA to target PRR15. These results suggest that PRR15 enhances trophoblast viability and survival during early implantation and placentation.
Collapse
Affiliation(s)
- Katherine C. Gates
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Lindsey N. Goetzmann
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Jeremy D. Cantlon
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Kimberly M. Jeckel
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
| | - Russell V. Anthony
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
41
|
Baker BC, Mackie FL, Lean SC, Greenwood SL, Heazell AEP, Forbes K, Jones RL. Placental dysfunction is associated with altered microRNA expression in pregnant women with low folate status. Mol Nutr Food Res 2017; 61. [PMID: 28105727 PMCID: PMC5573923 DOI: 10.1002/mnfr.201600646] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 11/27/2016] [Accepted: 01/10/2017] [Indexed: 12/21/2022]
Abstract
SCOPE Low maternal folate status during pregnancy increases the risk of delivering small for gestational age (SGA) infants, but the mechanistic link between maternal folate status, SGA, and placental dysfunction is unknown. microRNAs (miRNAs) are altered in pregnancy pathologies and by folate in other systems. We hypothesized that low maternal folate status causes placental dysfunction, mediated by altered miRNA expression. METHODS AND RESULTS A prospective observational study recruited pregnant adolescents and assessed third trimester folate status and placental function. miRNA array, QPCR, and bioinformatics identified placental miRNAs and target genes. Low maternal folate status is associated with higher incidence of SGA infants (28% versus 13%, p < 0.05) and placental dysfunction, including elevated trophoblast proliferation and apoptosis (p < 0.001), reduced amino acid transport (p < 0.01), and altered placental hormones (pregnancy-associated plasma protein A, progesterone, and human placental lactogen). miR-222-3p, miR-141-3p, and miR-34b-5p were upregulated by low folate status (p < 0.05). Bioinformatics predicted a gene network regulating cell turnover. Quantitative PCR demonstrated that key genes in this network (zinc finger E-box binding homeobox 2, v-myc myelocytomatosis viral oncogene homolog (avian), and cyclin-dependent kinase 6) were reduced (p < 0.05) in placentas with low maternal folate status. CONCLUSION This study supports that placental dysfunction contributes to impaired fetal growth in women with low folate status and suggests altered placental expression of folate-sensitive miRNAs and target genes as a mechanistic link.
Collapse
Affiliation(s)
- Bernadette C Baker
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | - Fiona L Mackie
- Centre of Women's and Newborn's Health & Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Samantha C Lean
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | - Susan L Greenwood
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | | | - Karen Forbes
- Division of Reproduction and Early Development, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Rebecca L Jones
- Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
42
|
Moindjie H, Santos ED, Gouesse RJ, Swierkowski-Blanchard N, Serazin V, Barnea ER, Vialard F, Dieudonné MN. Preimplantation factor is an anti-apoptotic effector in human trophoblasts involving p53 signaling pathway. Cell Death Dis 2016; 7:e2504. [PMID: 27906186 PMCID: PMC5261002 DOI: 10.1038/cddis.2016.382] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 09/02/2016] [Accepted: 10/11/2016] [Indexed: 12/13/2022]
Abstract
From the earliest stages of gestation, embryonic-maternal interaction has a key role in a successful pregnancy. Various factors present during gestation may significantly influence this type of juxta/paracrine interaction. PreImplantation Factor (PIF) is a recently identified factor with activity at the fetomaternal interface. PIF is secreted by viable embryos and directly controls placental development by increasing the invasive capacity of human extravillous trophoblasts (EVTs). To further specify PIF's role in the human placenta, we analyzed the genome-wide expression profile of the EVT in the presence of a synthetic PIF analog (sPIF). We found that sPIF exposure altered several pathways related to p53 signaling, survival and the immune response. Functional assays revealed that sPIF acts through the p53 pathway to reduce both early and late trophoblast apoptosis. More precisely, sPIF (i) decreases the phosphorylation of p53 at Ser-15, (ii) enhances the B-cell lymphoma-2 (BCL2) expression and (iii) reduces the BCL2-associated X protein (BAX) and BCL2 homologous antagonist killer (BAK) mRNA expression levels. Furthermore, invalidation experiments of TP53 allowed us to demonstrate that PIF's effects on placental apoptosis seemed to be essentially mediated by this gene. We have clearly shown that p53 and sPIF pathways could interact in human trophoblast and thus promotes cell survival. Furthermore, sPIF was found to regulate a gene network related to immune tolerance in the EVT, which emphasizes the beneficial effect of this peptide on the human placenta. Finally, the PIF protein levels in placentas from pregnancies affected by preeclampsia or intra-uterine growth restriction were significantly lower than in gestational age-matched control placentas. Taken as a whole, our results suggest that sPIF protects the EVT's functional status through a variety of mechanisms. Clinical application of sPIF in the treatment of disorders of early pregnancy can be envisioned.
Collapse
Affiliation(s)
- Hadia Moindjie
- GIG-EA7404, Université de Versailles Saint-Quentin-en-Yvelines - Paris Saclay, Unité de Formation et de Recherche des Sciences de la Santé-Simone Veil, Montigny-le Bretonneux, France
| | - Esther Dos Santos
- GIG-EA7404, Université de Versailles Saint-Quentin-en-Yvelines - Paris Saclay, Unité de Formation et de Recherche des Sciences de la Santé-Simone Veil, Montigny-le Bretonneux, France.,Service de Biologie Médicale, Centre Hospitalier de Poissy-Saint Germain, Poissy, France
| | - Rita-Josiane Gouesse
- GIG-EA7404, Université de Versailles Saint-Quentin-en-Yvelines - Paris Saclay, Unité de Formation et de Recherche des Sciences de la Santé-Simone Veil, Montigny-le Bretonneux, France
| | - Nelly Swierkowski-Blanchard
- Département de Biologie de la Reproduction, Cytogénétique, Gynécologie et Obstétrique, Centre Hospitalier de Poissy-Saint Germain, Poissy, France
| | - Valérie Serazin
- GIG-EA7404, Université de Versailles Saint-Quentin-en-Yvelines - Paris Saclay, Unité de Formation et de Recherche des Sciences de la Santé-Simone Veil, Montigny-le Bretonneux, France.,Service de Biologie Médicale, Centre Hospitalier de Poissy-Saint Germain, Poissy, France
| | - Eytan R Barnea
- Society for the Investigation of Early Pregnancy, Cherry Hill, NJ, USA.,BioIncept, LLC, Cherry Hill, NJ, USA
| | - François Vialard
- GIG-EA7404, Université de Versailles Saint-Quentin-en-Yvelines - Paris Saclay, Unité de Formation et de Recherche des Sciences de la Santé-Simone Veil, Montigny-le Bretonneux, France.,Département de Biologie de la Reproduction, Cytogénétique, Gynécologie et Obstétrique, Centre Hospitalier de Poissy-Saint Germain, Poissy, France
| | - Marie-Noëlle Dieudonné
- GIG-EA7404, Université de Versailles Saint-Quentin-en-Yvelines - Paris Saclay, Unité de Formation et de Recherche des Sciences de la Santé-Simone Veil, Montigny-le Bretonneux, France
| |
Collapse
|
43
|
|
44
|
Carbenoxolone exposure during late gestation in rats alters placental expressions of p53 and estrogen receptors. Eur J Pharmacol 2016; 791:675-685. [PMID: 27693517 DOI: 10.1016/j.ejphar.2016.09.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/20/2016] [Accepted: 09/27/2016] [Indexed: 12/31/2022]
Abstract
Gestational carbenoxolone exposure inhibits placental 11β-hydroxysteroid dehydrogenase (11β-HSD), the physiological barrier for glucocorticoids, which increases fetal exposure to glucocorticoids and induces intrauterine growth restriction (IUGR). We hypothesized that carbenoxolone exposure influences the expression of placental estrogen receptors-α and β (ERα & ERβ) and p53 leading to inhibited fetal and placental growth. Pregnant Sprague-Dawley rats were injected twice daily with either carbenoxolone (10mg/kg; s.c.) or vehicle (control group) from gestational days (dg) 12 onwards. Maternal blood and placentas were collected on 16 dg, 19 dg and 21 dg. The expression of ERα, ERβ and p53 were studied in placental basal and labyrinth zones by RT-PCR, Western blotting and immunohistochemistry. Carbenoxolone did not affect placental and fetal body weights, but ELISA showed decreased estradiol levels on 19 dg and 21 dg, and increased maternal luteinizing hormone levels on all dg. The follicle stimulating hormone levels decreased on 16 dg and 19 dg, and increased on 21 dg. Carbenoxolone decreased ERα mRNA levels on 16 dg in both zones and its protein level on 19 dg in the labyrinth zone. However, carbenoxolone increased ERβ mRNA levels on 19 dg and 21 dg and protein levels on 16 dg and 19 dg in the labyrinth zone. The p53 mRNA levels increased on all dg, but its protein levels increased on 21 dg in both zones. In conclusion, carbenoxolone exposure changes placental p53, ERα, ERβ expression in favor of cell death but these changes do not induce IUGR in rats.
Collapse
|
45
|
Paixão ES, Teixeira MG, Costa MDCN, Rodrigues LC. Dengue during pregnancy and adverse fetal outcomes: a systematic review and meta-analysis. THE LANCET. INFECTIOUS DISEASES 2016; 16:857-865. [DOI: 10.1016/s1473-3099(16)00088-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/04/2016] [Accepted: 02/05/2016] [Indexed: 10/22/2022]
|
46
|
Pérez-Pérez A, Toro AR, Vilarino-Garcia T, Guadix P, Maymó JL, Dueñas JL, Varone CL, Sánchez-Margalet V. Leptin reduces apoptosis triggered by high temperature in human placental villous explants: The role of the p53 pathway. Placenta 2016; 42:106-113. [PMID: 27238720 DOI: 10.1016/j.placenta.2016.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 03/17/2016] [Accepted: 03/20/2016] [Indexed: 11/21/2022]
Abstract
Maternal fever is common during pregnancy and has for many years been suspected to harm the developing fetus. Whether increased maternal temperature produces exaggerated apoptosis in trophoblast cells remains unclear. Since p53 is a critical regulator of apoptosis we hypothesized that increased temperature in placenta produces abnormal expression of proteins in the p53 pathway and finally caspase-3 activation. Moreover, leptin, produced by placenta, is known to promote the proliferation and survival of trophoblastic cells. Thus, we aimed to study the possible role of leptin preventing apoptosis triggered by high temperature, as well as the molecular mechanisms underlying this effect. Fresh placental tissue was collected from normal pregnancies. Explants of placental villi were exposed to 37 °C, 40 °C and 42 °C during 3 h in the presence or absence of 10 nM leptin in DMEM-F12 medium. Western blotting and qRT-PCR was performed to analyze the expression of p53 and downstream effector, P53AIP1, Mdm2, p21, BAX and BCL-2 as well as the activated cleaved form of caspase-3 and the fragment of cytokeratin-18 (CK-18) cleaved at Asp396 (neoepitope M30). Phosphorylation of the Ser 46 residue on p53, the expression of P53AIP1, Mdm2, p21, as well as caspase-3 and CK-18 were significantly increased in explants at 40 °C and 42 °C. Conversely, these effects were significantly attenuated by leptin 10 nM at both 40 °C and 42 °C. The BCL2/BAX ratio was also significantly decreased in explants at 40 °C and 42 °C compared with explants incubated at 37 °C, which was prevented by leptin stimulation. These data illustrate the potential role of leptin for reducing apoptosis in trophoblast explants, including trophoblastic cells, triggered by high temperature, by preventing the activation of p53 signaling.
Collapse
Affiliation(s)
- Antonio Pérez-Pérez
- Department of Medical Biochemistry and Molecular Biology, Virgen Macarena University Hospital, University of Seville, Spain
| | - Ayelén R Toro
- Department of Biological Chemistry, School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Teresa Vilarino-Garcia
- Department of Medical Biochemistry and Molecular Biology, Virgen Macarena University Hospital, University of Seville, Spain
| | - Pilar Guadix
- Department of Obstetrics and Gynecology, Virgen Macarena University Hospital, University of Seville, Spain
| | - Julieta L Maymó
- Department of Biological Chemistry, School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - José L Dueñas
- Department of Obstetrics and Gynecology, Virgen Macarena University Hospital, University of Seville, Spain
| | - Cecilia L Varone
- Department of Biological Chemistry, School of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Víctor Sánchez-Margalet
- Department of Medical Biochemistry and Molecular Biology, Virgen Macarena University Hospital, University of Seville, Spain.
| |
Collapse
|
47
|
Londero AP, Orsaria M, Marzinotto S, Grassi T, Fruscalzo A, Calcagno A, Bertozzi S, Nardini N, Stella E, Lellé RJ, Driul L, Tell G, Mariuzzi L. Placental aging and oxidation damage in a tissue micro-array model: an immunohistochemistry study. Histochem Cell Biol 2016; 146:191-204. [PMID: 27106773 DOI: 10.1007/s00418-016-1435-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2016] [Indexed: 12/12/2022]
Abstract
To evaluate the expression of markers correlated with cellular senescence and DNA damage (8-hydroxy-2'-deoxy-guanosine (8-OHdG), p53, p21, APE1/Ref-1 (APE1), interleukin (IL-6 and IL-8) in placentas from healthy and pathologic pregnancies. This retrospective study considered a placental tissue micro-array containing 92 controls from different gestational ages and 158 pathological cases including preeclampsia (PE), HELLP syndrome (hemolysis, elevated liver enzymes, low platelet count), small for gestational age (SGA) fetuses, and intrauterine growth restriction (IUGR) occurring at different gestational ages. In this study, we demonstrated a significant influence of gestational age on the expression in the trophoblast of 8-OHdG, p53, p21, APE1, and IL-6. In placentas of cases affected by PE, HELLP, or IUGR, there was an increased expression of 8-OHdG, p53, APE1, and IL-6 compared to controls (only IL-8 was significantly decreased in cases). In both groups of pathology between 22- and 34-week gestation and after 34-week gestation, APE1 levels were higher in the trophoblast of women affected by hypertensive disorders of pregnancy than women carrying an IUGR fetus. The cytoplasmic expression of 8-OHdG was increased in placentas in IUGR cases compared to PE or HELLP pregnancies. In cases after 34-week gestation, p21 was higher in SGA and IUGR than in controls and late PE. Moreover, p53 was increased after 34-week gestation in IUGR pregnancies. Placentas from pathological pregnancies had an altered expression of 8-OHdG, p53, p21, APE1, IL-6, and IL-8. The alterations of intracellular pathways involving these elements may be the cause or the consequence of placental dysfunction, but in any case reflect an impaired placental function, possibly due to increased aging velocity in pathologic cases.
Collapse
Affiliation(s)
- Ambrogio P Londero
- Clinic of Obstetrics and Gynecology, Deparment of Experimental Clinical and Medical Science, University of Udine, Piazzale SM della Misericordia, 15, 33100, Udine, Italy. .,Unit of Obstetrics and Gynecology, S. Polo Hospital, 34074, Monfalcone, GO, Italy.
| | - Maria Orsaria
- Department of Medical and Biological Sciences, University of Udine, 33100, Udine, Italy
| | - Stefania Marzinotto
- Department of Medical and Biological Sciences, University of Udine, 33100, Udine, Italy
| | - Tiziana Grassi
- Clinic of Obstetrics and Gynecology, Deparment of Experimental Clinical and Medical Science, University of Udine, Piazzale SM della Misericordia, 15, 33100, Udine, Italy
| | - Arrigo Fruscalzo
- Frauenklinik, St Franziskus Hospital, Münster, Germany.,Clinic of Obstetrics and Gynecology and Institute of Pathology, University Hospital of Münster, Albert-Schweitzer-Campus 1, Gebäude: A1, 48149, Münster, Germany
| | - Angelo Calcagno
- Clinic of Obstetrics and Gynecology, Deparment of Experimental Clinical and Medical Science, University of Udine, Piazzale SM della Misericordia, 15, 33100, Udine, Italy
| | - Serena Bertozzi
- Department of Surgical Oncology, IRCCS CRO, 33081, Aviano, PN, Italy
| | - Nastassia Nardini
- Department of Medical and Biological Sciences, University of Udine, 33100, Udine, Italy
| | - Enrica Stella
- Clinic of Obstetrics and Gynecology, Deparment of Experimental Clinical and Medical Science, University of Udine, Piazzale SM della Misericordia, 15, 33100, Udine, Italy
| | - Ralph J Lellé
- Clinic of Obstetrics and Gynecology and Institute of Pathology, University Hospital of Münster, Albert-Schweitzer-Campus 1, Gebäude: A1, 48149, Münster, Germany
| | - Lorenza Driul
- Clinic of Obstetrics and Gynecology, Deparment of Experimental Clinical and Medical Science, University of Udine, Piazzale SM della Misericordia, 15, 33100, Udine, Italy
| | - Gianluca Tell
- Department of Medical and Biological Sciences, University of Udine, 33100, Udine, Italy
| | - Laura Mariuzzi
- Department of Medical and Biological Sciences, University of Udine, 33100, Udine, Italy
| |
Collapse
|
48
|
Ptacek I, Smith A, Garrod A, Bullough S, Bradley N, Batra G, Sibley CP, Jones RL, Brownbill P, Heazell AEP. Quantitative assessment of placental morphology may identify specific causes of stillbirth. BMC Clin Pathol 2016; 16:1. [PMID: 26865834 PMCID: PMC4748636 DOI: 10.1186/s12907-016-0023-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 02/04/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Stillbirth is frequently the result of pathological processes involving the placenta. Understanding the significance of specific lesions is hindered by qualitative subjective evaluation. We hypothesised that quantitative assessment of placental morphology would identify alterations between different causes of stillbirth and that placental phenotype would be independent of post-mortem effects and differ between live births and stillbirths with the same condition. METHODS Placental tissue was obtained from stillbirths with an established cause of death, those of unknown cause and live births. Image analysis was used to quantify different facets of placental structure including: syncytial nuclear aggregates (SNAs), proliferative cells, blood vessels, leukocytes and trophoblast area. These analyses were then applied to placental tissue from live births and stillbirths associated with fetal growth restriction (FGR), and to placental lobules before and after perfusion of the maternal side of the placental circulation to model post-mortem effects. RESULTS Different causes of stillbirth, particularly FGR, cord accident and hypertension had altered placental morphology compared to healthy live births. FGR stillbirths had increased SNAs and trophoblast area and reduced proliferation and villous vascularity; 2 out of 10 stillbirths of unknown cause had similar placental morphology to FGR. Stillbirths with FGR had reduced vascularity, proliferation and trophoblast area compared to FGR live births. Ex vivo perfusion did not reproduce the morphological findings of stillbirth. CONCLUSION These preliminary data suggest that addition of quantitative assessment of placental morphology may distinguish between different causes of stillbirth; these changes do not appear to be due to post-mortem effects. Applying quantitative assessment in addition to qualitative assessment might reduce the proportion of unexplained stillbirths.
Collapse
Affiliation(s)
- Imogen Ptacek
- />Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Oxford Rd, Manchester, M13 9PL UK
- />Maternal and Fetal Health Research Centre, 5th floor (Research), St Mary’s Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Anna Smith
- />Department of Histopathology, Royal Manchester Children’s Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL UK
| | - Ainslie Garrod
- />Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Oxford Rd, Manchester, M13 9PL UK
- />Maternal and Fetal Health Research Centre, 5th floor (Research), St Mary’s Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Sian Bullough
- />Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Oxford Rd, Manchester, M13 9PL UK
- />Maternal and Fetal Health Research Centre, 5th floor (Research), St Mary’s Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Nicola Bradley
- />Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Oxford Rd, Manchester, M13 9PL UK
- />Maternal and Fetal Health Research Centre, 5th floor (Research), St Mary’s Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Gauri Batra
- />Department of Histopathology, Royal Manchester Children’s Hospital, Central Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL UK
| | - Colin P. Sibley
- />Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Oxford Rd, Manchester, M13 9PL UK
- />Maternal and Fetal Health Research Centre, 5th floor (Research), St Mary’s Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Rebecca L. Jones
- />Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Oxford Rd, Manchester, M13 9PL UK
- />Maternal and Fetal Health Research Centre, 5th floor (Research), St Mary’s Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Paul Brownbill
- />Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Oxford Rd, Manchester, M13 9PL UK
- />Maternal and Fetal Health Research Centre, 5th floor (Research), St Mary’s Hospital, Oxford Road, Manchester, M13 9WL UK
| | - Alexander E. P. Heazell
- />Institute of Human Development, Faculty of Medical and Human Sciences, University of Manchester, Oxford Rd, Manchester, M13 9PL UK
- />Maternal and Fetal Health Research Centre, 5th floor (Research), St Mary’s Hospital, Oxford Road, Manchester, M13 9WL UK
| |
Collapse
|
49
|
Increased Apoptosis, Altered Oxygen Signaling, and Antioxidant Defenses in First-Trimester Pregnancies with High-Resistance Uterine Artery Blood Flow. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2731-41. [DOI: 10.1016/j.ajpath.2015.06.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 05/22/2015] [Accepted: 06/01/2015] [Indexed: 12/29/2022]
|
50
|
|