1
|
Shi X, Xi C, Dong B, Yan Z, Liu W, Gao S, Chen D. Maternal infection with SARS-CoV-2 during early pregnancy induces hypoxia at the maternal-fetal interface. Cell Prolif 2025; 58:e13749. [PMID: 39375953 PMCID: PMC11839197 DOI: 10.1111/cpr.13749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/31/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic increases the risk of adverse fetal outcomes during pregnancy. Maternal infection during pregnancy, particularly with cytomegalovirus (CMV), hepatitis B and C virus, and human immunodeficiency virus can have detrimental effects on both mother and fetus, potentially leading to adverse outcomes such as spontaneous abortion or neonatal infection. However, the impact of severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection on the maternal-fetal interface remains poorly understood. In this study, we initially utilised immunofluorescence and immunohistochemical to investigate placental samples from pregnant women who were infected with SARS-CoV-2 during the first trimester. Our data indicate that infection in the first trimester induces an upregulation of hypoxia inducible factor (HIF) levels at the maternal-fetal interface. Subsequently, single-cell RNA sequencing and metabolomics sequencing analyses reveal alterations in maternal-fetal interface. Remarkably, immune cells exhibited low expression levels of HIF possibly associated with immune activation. Furthermore, our findings demonstrate a gradual reduction in transcriptome and metabolic changes as gestation progressed beyond 12-16 weeks compared to samples obtained at 6-8 weeks gestation. Overall, our study suggests that early-stage SARS-CoV-2 infection during the first trimester leads to severe hypoxia and aberrant cell metabolism at the maternal-fetal interface which gradually resolves as pregnancy progresses. Nevertheless, these abnormal changes may have long-term implications for maternal-fetal interface development.
Collapse
Affiliation(s)
- Xiaohui Shi
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangChina
| | - Chenxiang Xi
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Baoxing Dong
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Zihui Yan
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Wenqiang Liu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Shaorong Gao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Frontier Science Center for Stem Cell Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Di Chen
- Center for Reproductive Medicine of The Second Affiliated Hospital, Center for Regeneration and Cell Therapy of Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute), Zhejiang University School of MedicineZhejiang UniversityHangzhouZhejiangChina
- Dr. Li Dak Sum and Yip Yio Chin Center for Stem Cell and Regenerative MedicineZhejiang UniversityHangzhouChina
- State Key Laboratory of Biobased Transportation Fuel TechnologyZhejiang UniversityHainingZhejiangChina
| |
Collapse
|
2
|
Lai H, Yang Y, Zhang J. Advances in post-translational modifications and recurrent spontaneous abortion. Gene 2024; 927:148700. [PMID: 38880188 DOI: 10.1016/j.gene.2024.148700] [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: 01/31/2024] [Revised: 05/25/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Recurrent spontaneous abortion (RSA) is defined as two or more pregnancy loss, which affects approximately 1-2% of women's fertility. The etiology of RSA has not yet been fully revealed, which poses a great problem for clinical treatment. Post- translational modifications(PTMs) are chemical modifications that play a crucial role in the functional proteome. A considerable number of published studies have shown the relationship between post-translational modifications of various proteins and RSA. The study of PTMs contributes to elucidating the role of modified proteins in the pathogenesis of RSA, as well as the design of more effective diagnostic/prognostic tools and more targeted treatments. Most reviews in the field of RSA have only focused on RNA epigenomics research. The present review reports the latest research developments of PTMs related to RSA, such as glycosylation, phosphorylation, Methylation, Acetylation, Ubiquitination, etc.
Collapse
Affiliation(s)
- Hanhong Lai
- Jinan University, Guangzhou, Guangdong 510632, People's Republic of China
| | - Yi Yang
- Jinan University, Guangzhou, Guangdong 510632, People's Republic of China
| | - Jun Zhang
- Jinan University, Guangzhou, Guangdong 510632, People's Republic of China.
| |
Collapse
|
3
|
Bowman-Gibson S, Chandiramani C, Stone ML, Waker CA, Rackett TM, Maxwell RA, Dhanraj DN, Brown TL. Streamlined Analysis of Maternal Plasma Indicates Small Extracellular Vesicles are Significantly Elevated in Early-Onset Preeclampsia. Reprod Sci 2024; 31:2771-2782. [PMID: 38777947 PMCID: PMC11393201 DOI: 10.1007/s43032-024-01591-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Preeclampsia (PE) is a leading cause of maternal and fetal mortality and morbidity. While placental dysfunction is a core underlying issue, the pathogenesis of this disorder is thought to differ between early-onset (EOPE) and late-onset (LOPE) subtypes. As recent reports suggest that small extracellular vesicles (sEVs) contribute to the development of PE, we have compared systemic sEV concentrations between normotensive, EOPE, and LOPE pregnancies. To circumvent lengthy isolation techniques and intermediate filtration steps, a streamlined approach was developed to evaluate circulating plasma sEVs from maternal plasma. Polymer-based precipitation and purification were used to isolate total systemic circulating maternal sEVs, free from bias toward specific surface marker expression or extensive subpurification. Immediate Nanoparticle Tracking Analysis (NTA) of freshly isolated sEV samples afforded a comprehensive analysis that can be completed within hours, avoiding confounding freeze-thaw effects of particle aggregation and degradation.Rather than exosomal subpopulations, our findings indicate a significant elevation in the total number of circulating maternal sEVs in patients with EOPE. This streamlined approach also preserves sEV-bound protein and microRNA (miRNA) that can be used for potential biomarker analysis. This study is one of the first to demonstrate that maternal plasma sEVs harbor full-length hypoxia inducible factor 1 alpha (HIF-1α) protein, with EOPE sEVs carrying higher levels of HIF-1α compared to control sEVs. The detection of HIF-1α and its direct signaling partner microRNA-210 (miR-210) within systemic maternal sEVs lays the groundwork for identifying how sEV signaling contributes to the development of preeclampsia. When taken together, our quantitative and qualitative results provide compelling evidence to support the translational potential of streamlined sEV analysis for future use in the clinical management of patients with EOPE.
Collapse
Affiliation(s)
- Scout Bowman-Gibson
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA
| | - Chandni Chandiramani
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - Madison L Stone
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA
| | - Christopher A Waker
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA
| | - Traci M Rackett
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - Rose A Maxwell
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - David N Dhanraj
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA
| | - Thomas L Brown
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, 3640 Colonel Glenn Highway, 457 NEC Building, Dayton, OH, 45435, USA.
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, 45435, USA.
| |
Collapse
|
4
|
de Lima Castro M, Dos Passos RR, Justina VD, do Amaral WN, Giachini FR. Physiological and pathological evidence of O-GlcNAcylation regulation during pregnancy related process. Placenta 2023; 141:43-50. [PMID: 37210277 DOI: 10.1016/j.placenta.2023.04.018] [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: 01/23/2023] [Revised: 03/22/2023] [Accepted: 04/25/2023] [Indexed: 05/22/2023]
Abstract
O-GlcNAcylation is a dynamic and reversible post-translational modification (PTM) controlled by the enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Changes in its expression lead to a breakdown in cellular homeostasis, which is linked to several pathological processes. Placentation and embryonic development are periods of high cell activity, and imbalances in cell signaling pathways can result in infertility, miscarriage, or pregnancy complications. O-GlcNAcylation is involved in cellular processes such as genome maintenance, epigenetic regulation, protein synthesis/degradation, metabolic pathways, signaling pathways, apoptosis, and stress response. Trophoblastic differentiation/invasion and placental vasculogenesis, as well as zygote viability and embryonic neuronal development, are all dependent on O-GlcNAcylation. This PTM is required for pluripotency, which is a required condition for embryonic development. Further, this pathway is a nutritional sensor and cell stress marker, which is primarily measured by the OGT enzyme and its product, protein O-GlcNAcylation. Yet, this post-translational modification is enrolled in metabolic and cardiovascular adaptations during pregnancy. Finally, evidence of how O-GlcNAc impacts pregnancy during pathological conditions such as hyperglycemia, gestational diabetes, hypertension, and stress disorders are reviewed. Considering this scenario, progress in understanding the role of O- GlcNAcylation in pregnancy is required.
Collapse
Affiliation(s)
- Marta de Lima Castro
- Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goias, Goiânia, Brazil
| | - Rinaldo Rodrigues Dos Passos
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Vanessa Dela Justina
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Waldemar Naves do Amaral
- Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goias, Goiânia, Brazil
| | - Fernanda Regina Giachini
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil.
| |
Collapse
|
5
|
Pintye D, Sziva RE, Mastyugin M, Young BC, Jacas S, Török M, Salahuddin S, Jagtap P, Southan GJ, Zsengellér ZK. A Novel Dual-Function Redox Modulator Relieves Oxidative Stress and Anti-Angiogenic Response in Placental Villus Explant Exposed to Hypoxia-Relevance for Preeclampsia Therapy. BIOLOGY 2023; 12:1229. [PMID: 37759628 PMCID: PMC10525611 DOI: 10.3390/biology12091229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Preeclampsia (PE) is a severe, life-threatening complication during pregnancy (~5-7%), and no causative treatment is available. Early aberrant spiral artery remodeling is associated with placental stress and the release of oxygen radicals and other reactive oxygen species (ROS) in the placenta. This precedes the production of anti-angiogenic factors, which ultimately leads to endothelial and trophoblast damage and the key features of PE. We tested whether a novel dual-function redox modulator-AKT-1005-can effectively reduce placental oxidative stress and alleviate PE symptoms in vitro. METHOD Isolated human villous explants were exposed to hypoxia and assessed to determine whether improving cell-redox function with AKT-1005 diminished ROS production, mitochondrial stress, production of the transcription factor HIF1A, and downstream anti-angiogenic responses (i.e., sFLT1, sEng production). MitoTEMPO was used as a reference antioxidant. RESULTS In our villous explant assays, pretreatment with AKT-1005 reduced mitochondrial-derived ROS production, reduced HIF-1A, sFLT1, and sEng protein expression, while increasing VEGF in hypoxia-exposed villous trophoblast cells, with better efficiency than MitoTEMPO. In addition, AKT-1005 improved mitochondrial electron chain enzyme activity in the stressed explant culture. CONCLUSIONS The redox modulator AKT-1005 has the potential to intervene with oxidative stress and can be efficacious for PE therapy. Future studies are underway to assess the in vivo efficacy of HMP.
Collapse
Affiliation(s)
- Diana Pintye
- Department of Medicine, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (D.P.); (S.J.)
| | - Réka E. Sziva
- Department of Medicine, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (D.P.); (S.J.)
- Department of Obstetrics and Gynecology, Semmelweis University, 1082 Budapest, Hungary
| | - Maxim Mastyugin
- Department of Chemistry, University of Massachusetts, Boston, MA 02125, USA; (M.M.); (M.T.)
| | - Brett C. Young
- Department of Obstetrics and Gynecology, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (B.C.Y.); (S.S.)
| | - Sonako Jacas
- Department of Medicine, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (D.P.); (S.J.)
| | - Marianna Török
- Department of Chemistry, University of Massachusetts, Boston, MA 02125, USA; (M.M.); (M.T.)
| | - Saira Salahuddin
- Department of Obstetrics and Gynecology, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (B.C.Y.); (S.S.)
| | | | | | - Zsuzsanna K. Zsengellér
- Department of Medicine, Beth Israel Lahey Health, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (D.P.); (S.J.)
| |
Collapse
|
6
|
Bueno-Sánchez JC, Gómez-Gutiérrez AM, Maldonado-Estrada JG, Quintana-Castillo JC. Expression of placental glycans and its role in regulating peripheral blood NK cells during preeclampsia: a perspective. Front Endocrinol (Lausanne) 2023; 14:1087845. [PMID: 37206444 PMCID: PMC10190602 DOI: 10.3389/fendo.2023.1087845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/03/2023] [Indexed: 05/21/2023] Open
Abstract
Preeclampsia is a pregnancy-related multisystem disorder characterized by altered trophoblast invasion, oxidative stress, exacerbation of systemic inflammatory response, and endothelial damage. The pathogenesis includes hypertension and mild-to-severe microangiopathy in the kidney, liver, placenta, and brain. The main mechanisms involved in its pathogenesis have been proposed to limit trophoblast invasion and increase the release of extracellular vesicles from the syncytiotrophoblast into the maternal circulation, exacerbating the systemic inflammatory response. The placenta expresses glycans as part of its development and maternal immune tolerance during gestation. The expression profile of glycans at the maternal-fetal interface may play a fundamental role in physiological pregnancy changes and disorders such as preeclampsia. It is unclear whether glycans and their lectin-like receptors are involved in the mechanisms of maternal-fetal recognition by immune cells during pregnancy homeostasis. The expression profile of glycans appears to be altered in hypertensive disorders of pregnancy, which could lead to alterations in the placental microenvironment and vascular endothelium in pregnancy conditions such as preeclampsia. Glycans with immunomodulatory properties at the maternal-fetal interface are altered in early-onset severe preeclampsia, implying that innate immune system components, such as NK cells, exacerbate the systemic inflammatory response observed in preeclampsia. In this article, we discuss the evidence for the role of glycans in gestational physiology and the perspective of glycobiology on the pathophysiology of hypertensive disorders in gestation.
Collapse
Affiliation(s)
- Julio C. Bueno-Sánchez
- Reproduction Group, Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Medellín, Colombia
- Department of Obstetrics and Gynecology, School of Medicine, Universidad de Antioquia, Medellín, Colombia
- Red Iberoamericana de Alteraciones Vasculares en Trastornos del Embarazo (RIVATREM), Chillan, Chile
| | - Alejandra M. Gómez-Gutiérrez
- Reproduction Group, Department of Physiology and Biochemistry, School of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Juan G. Maldonado-Estrada
- One Health and Veterinary Innovative Research & Development (OHVRI) Research Group, Escuela de Medicina Veterinaria, Universidad de Antioquia, Medellín, Colombia
| | | |
Collapse
|
7
|
Zhou H, Zhao C, Wang P, Yang W, Zhu H, Zhang S. Regulators involved in trophoblast syncytialization in the placenta of intrauterine growth restriction. Front Endocrinol (Lausanne) 2023; 14:1107182. [PMID: 36798658 PMCID: PMC9927020 DOI: 10.3389/fendo.2023.1107182] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Placental dysfunction refers to the insufficiency of placental perfusion and chronic hypoxia during early pregnancy, which impairs placental function and causes inadequate supply of oxygen and nutrients to the fetus, affecting fetal development and health. Fetal intrauterine growth restriction, one of the most common outcomes of pregnancy-induced hypertensions, can be caused by placental dysfunction, resulting from deficient trophoblast syncytialization, inadequate trophoblast invasion and impaired vascular remodeling. During placental development, cytotrophoblasts fuse to form a multinucleated syncytia barrier, which supplies oxygen and nutrients to meet the metabolic demands for fetal growth. A reduction in the cell fusion index and the number of nuclei in the syncytiotrophoblast are found in the placentas of pregnancies complicated by IUGR, suggesting that the occurrence of IUGR may be related to inadequate trophoblast syncytialization. During the multiple processes of trophoblasts syncytialization, specific proteins and several signaling pathways are involved in coordinating these events and regulating placental function. In addition, epigenetic modifications, cell metabolism, senescence, and autophagy are also involved. Study findings have indicated several abnormally expressed syncytialization-related proteins and signaling pathways in the placentas of pregnancies complicated by IUGR, suggesting that these elements may play a crucial role in the occurrence of IUGR. In this review, we discuss the regulators of trophoblast syncytialization and their abnormal expression in the placentas of pregnancies complicated by IUGR.
Collapse
Affiliation(s)
- Hanjing Zhou
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Chenqiong Zhao
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Peixin Wang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Weijie Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Haiyan Zhu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- *Correspondence: Songying Zhang, ; Haiyan Zhu,
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- *Correspondence: Songying Zhang, ; Haiyan Zhu,
| |
Collapse
|
8
|
Sallais J, Park C, Alahari S, Porter T, Liu R, Kurt M, Farrell A, Post M, Caniggia I. HIF1 inhibitor acriflavine rescues early-onset preeclampsia phenotype in mice lacking placental prolyl hydroxylase domain protein 2. JCI Insight 2022; 7:158908. [PMID: 36227697 PMCID: PMC9746916 DOI: 10.1172/jci.insight.158908] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/29/2022] [Indexed: 01/12/2023] Open
Abstract
Preeclampsia is a serious pregnancy disorder that lacks effective treatments other than delivery. Improper sensing of oxygen changes during placentation by prolyl hydroxylases (PHDs), specifically PHD2, causes placental hypoxia-inducible factor-1 (HIF1) buildup and abnormal downstream signaling in early-onset preeclampsia, yet therapeutic targeting of HIF1 has never been attempted. Here we generated a conditional (placenta-specific) knockout of Phd2 in mice (Phd2-/- cKO) to reproduce HIF1 excess and to assess anti-HIF therapy. Conditional deletion of Phd2 in the junctional zone during pregnancy increased placental HIF1 content, resulting in abnormal placentation, impaired remodeling of the uterine spiral arteries, and fetal growth restriction. Pregnant dams developed new-onset hypertension at midgestation (E9.5) in addition to proteinuria and renal and cardiac pathology, hallmarks of severe preeclampsia in humans. Daily injection of acriflavine, a small molecule inhibitor of HIF1, to pregnant Phd2-/- cKO mice from E7.5 (prior to hypertension) or E10.5 (after hypertension had been established) to E14.5 corrected placental dysmorphologies and improved fetal growth. Moreover, it reduced maternal blood pressure and reverted renal and myocardial pathology. Thus, therapeutic targeting of the HIF pathway may improve placental development and function, as well as maternal and fetal health, in preeclampsia.
Collapse
Affiliation(s)
- Julien Sallais
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Institute of Medical Sciences, and
| | - Chanho Park
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada
| | - Sruthi Alahari
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada
| | - Tyler Porter
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Ruizhe Liu
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada
| | - Merve Kurt
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Abby Farrell
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Institute of Medical Sciences, and
| | - Martin Post
- Institute of Medical Sciences, and,Department of Physiology, University of Toronto, Ontario, Canada.,Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Institute of Medical Sciences, and,Department of Physiology, University of Toronto, Ontario, Canada.,Department of Obstetrics & Gynaecology, University of Toronto, Ontario, Canada
| |
Collapse
|
9
|
Garcés-Lázaro I, Kotzur R, Cerwenka A, Mandelboim O. NK Cells Under Hypoxia: The Two Faces of Vascularization in Tumor and Pregnancy. Front Immunol 2022; 13:924775. [PMID: 35769460 PMCID: PMC9234265 DOI: 10.3389/fimmu.2022.924775] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/17/2022] [Indexed: 01/14/2023] Open
Abstract
Environmental conditions greatly shape the phenotype and function of immune cells. Specifically, hypoxic conditions that exist within tissues and organs have been reported to affect both the adaptive and the innate immune system. Natural killer (NK) cells belong to the innate immune system. They are among the first immune cells responding to infections and are involved in tumor surveillance. NK cells produce cytokines that shape other innate and adaptive immune cells, and they produce cytolytic molecules leading to target cell killing. Therefore, they are not only involved in steady state tissue homeostasis, but also in pathogen and tumor clearance. Hence, understanding the role of NK cells in pathological and physiological immune biology is an emerging field. To date, it remains incompletely understood how the tissue microenvironment shapes NK cell phenotype and function. In particular, the impact of low oxygen concentrations in tissues on NK cell reactivity has not been systematically dissected. Here, we present a comprehensive review focusing on two highly compelling hypoxic tissue environments, the tumor microenvironment (pathological) and the decidua (physiological) and compare their impact on NK cell reactivity.
Collapse
Affiliation(s)
- Irene Garcés-Lázaro
- Department of Immunobiochemistry, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rebecca Kotzur
- The Lautenberg Center for General and Tumor Immunology, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
| | - Adelheid Cerwenka
- Department of Immunobiochemistry, Mannheim Institute of Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- *Correspondence: Adelheid Cerwenka, ; Ofer Mandelboim,
| | - Ofer Mandelboim
- The Lautenberg Center for General and Tumor Immunology, Institute for Medical Research Israel-Canada, The Hebrew University Hadassah Medical School, Jerusalem, Israel
- *Correspondence: Adelheid Cerwenka, ; Ofer Mandelboim,
| |
Collapse
|
10
|
McCracken SA, Seeho SKM, Carrodus T, Park JH, Woodland N, Gallery EDM, Morris JM, Ashton AW. Dysregulation of Oxygen Sensing/Response Pathways in Pregnancies Complicated by Idiopathic Intrauterine Growth Restriction and Early-Onset Preeclampsia. Int J Mol Sci 2022; 23:ijms23052772. [PMID: 35269911 PMCID: PMC8910827 DOI: 10.3390/ijms23052772] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 02/01/2023] Open
Abstract
Preeclampsia (PE) and intrauterine growth restriction (IUGR) are the leading causes of maternal and fetal morbidity/mortality. The central deficit in both conditions is impaired placentation due to poor trophoblast invasion, resulting in a hypoxic milieu in which oxidative stress contributes to the pathology. We examine the factors driving the hypoxic response in severely preterm PE (n = 19) and IUGR (n = 16) placentae compared to the spontaneous preterm (SPT) controls (n = 13) using immunoblotting, RT-PCR, immunohistochemistry, proximity ligation assays, and Co-IP. Both hypoxia-inducible factor (HIF)-1α and HIF-2α are increased at the protein level and functional in pathological placentae, as target genes prolyl hydroxylase domain (PHD)2, PHD3, and soluble fms-like tyrosine kinase-1 (sFlt-1) are increased. Accumulation of HIF-α-subunits occurs in the presence of accessory molecules required for their degradation (PHD1, PHD2, and PHD3 and the E3 ligase von Hippel–Lindau (VHL)), which were equally expressed or elevated in the placental lysates of PE and IUGR. However, complex formation between VHL and HIF-α-subunits is defective. This is associated with enhanced VHL/DJ1 complex formation in both PE and IUGR. In conclusion, we establish a significant mechanism driving the maladaptive responses to hypoxia in the placentae from severe PE and IUGR, which is central to the pathogenesis of both diseases.
Collapse
Affiliation(s)
- Sharon A. McCracken
- Division of Perinatal Medicine, Faculty of Medicine and Health, The University of Sydney, Northern Sydney Local Health District Research (Kolling Institute), St. Leonards, NSW 2065, Australia; (S.K.M.S.); (T.C.); (J.H.P.); (E.D.M.G.); (J.M.M.); (A.W.A.)
- Correspondence: ; Tel.: +612-9926-4832; Fax: +612-9926-5266
| | - Sean K. M. Seeho
- Division of Perinatal Medicine, Faculty of Medicine and Health, The University of Sydney, Northern Sydney Local Health District Research (Kolling Institute), St. Leonards, NSW 2065, Australia; (S.K.M.S.); (T.C.); (J.H.P.); (E.D.M.G.); (J.M.M.); (A.W.A.)
- Department of Obstetrics and Gynaecology, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - Tamara Carrodus
- Division of Perinatal Medicine, Faculty of Medicine and Health, The University of Sydney, Northern Sydney Local Health District Research (Kolling Institute), St. Leonards, NSW 2065, Australia; (S.K.M.S.); (T.C.); (J.H.P.); (E.D.M.G.); (J.M.M.); (A.W.A.)
- School of Biomedical Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Jenny H. Park
- Division of Perinatal Medicine, Faculty of Medicine and Health, The University of Sydney, Northern Sydney Local Health District Research (Kolling Institute), St. Leonards, NSW 2065, Australia; (S.K.M.S.); (T.C.); (J.H.P.); (E.D.M.G.); (J.M.M.); (A.W.A.)
| | - Narelle Woodland
- School of Biomedical Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia;
| | - Eileen D. M. Gallery
- Division of Perinatal Medicine, Faculty of Medicine and Health, The University of Sydney, Northern Sydney Local Health District Research (Kolling Institute), St. Leonards, NSW 2065, Australia; (S.K.M.S.); (T.C.); (J.H.P.); (E.D.M.G.); (J.M.M.); (A.W.A.)
- Department of Obstetrics and Gynaecology, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - Jonathan M. Morris
- Division of Perinatal Medicine, Faculty of Medicine and Health, The University of Sydney, Northern Sydney Local Health District Research (Kolling Institute), St. Leonards, NSW 2065, Australia; (S.K.M.S.); (T.C.); (J.H.P.); (E.D.M.G.); (J.M.M.); (A.W.A.)
- Department of Obstetrics and Gynaecology, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - Anthony W. Ashton
- Division of Perinatal Medicine, Faculty of Medicine and Health, The University of Sydney, Northern Sydney Local Health District Research (Kolling Institute), St. Leonards, NSW 2065, Australia; (S.K.M.S.); (T.C.); (J.H.P.); (E.D.M.G.); (J.M.M.); (A.W.A.)
- Department of Obstetrics and Gynaecology, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|
11
|
Amelio GS, Provitera L, Raffaeli G, Tripodi M, Amodeo I, Gulden S, Cortesi V, Manzoni F, Cervellini G, Tomaselli A, Pravatà V, Garrido F, Villamor E, Mosca F, Cavallaro G. Endothelial dysfunction in preterm infants: The hidden legacy of uteroplacental pathologies. Front Pediatr 2022; 10:1041919. [PMID: 36405831 PMCID: PMC9671930 DOI: 10.3389/fped.2022.1041919] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Millions of infants are born prematurely every year worldwide. Prematurity, particularly at lower gestational ages, is associated with high mortality and morbidity and is a significant global health burden. Pregnancy complications and preterm birth syndrome strongly impact neonatal clinical phenotypes and outcomes. The vascular endothelium is a pivotal regulator of fetal growth and development. In recent years, the key role of uteroplacental pathologies impairing endothelial homeostasis is emerging. Conditions leading to very and extremely preterm birth can be classified into two main pathophysiological patterns or endotypes: infection/inflammation and dysfunctional placentation. The first is frequently related to chorioamnionitis, whereas the second is commonly associated with hypertensive disorders of pregnancy and fetal growth restriction. The nature, timing, and extent of prenatal noxa may alter fetal and neonatal endothelial phenotype and functions. Changes in the luminal surface, oxidative stress, growth factors imbalance, and dysregulation of permeability and vascular tone are the leading causes of endothelial dysfunction in preterm infants. However, the available evidence regarding endothelial physiology and damage is limited in neonates compared to adults. Herein, we discuss the current knowledge on endothelial dysfunction in the infectious/inflammatory and dysfunctional placentation endotypes of prematurity, summarizing their molecular features, available biomarkers, and clinical impact. Furthermore, knowledge gaps, shadows, and future research perspectives are highlighted.
Collapse
Affiliation(s)
- Giacomo Simeone Amelio
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Livia Provitera
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Genny Raffaeli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Matteo Tripodi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ilaria Amodeo
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Silvia Gulden
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valeria Cortesi
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Francesca Manzoni
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Gaia Cervellini
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Andrea Tomaselli
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Valentina Pravatà
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Felipe Garrido
- Department of Pediatrics, Clínica Universidad de Navarra, Madrid, Spain
| | - Eduardo Villamor
- Department of Pediatrics, Maastricht University Medical Center (MUMC+), School for Oncology and Reproduction (GROW), University of Maastricht, Maastricht, Netherlands
| | - Fabio Mosca
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.,Department of Clinical Sciences and Community Health, Università Degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- Neonatal Intensive Care Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
12
|
Kornacki J, Gutaj P, Kalantarova A, Sibiak R, Jankowski M, Wender-Ozegowska E. Endothelial Dysfunction in Pregnancy Complications. Biomedicines 2021; 9:1756. [PMID: 34944571 PMCID: PMC8698592 DOI: 10.3390/biomedicines9121756] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/29/2022] Open
Abstract
The endothelium, which constitutes the inner layer of blood vessels and lymphatic structures, plays an important role in various physiological functions. Alterations in structure, integrity and function of the endothelial layer during pregnancy have been associated with numerous gestational complications, including clinically significant disorders, such as preeclampsia, fetal growth restriction, and diabetes. While numerous experimental studies have focused on establishing the role of endothelial dysfunction in pathophysiology of these gestational complications, their mechanisms remain unknown. Numerous biomarkers of endothelial dysfunction have been proposed, together with the mechanisms by which they relate to individual gestational complications. However, more studies are required to determine clinically relevant markers specific to a gestational complication of interest, as currently most of them present a significant overlap. Although the independent diagnostic value of such markers remains to be insufficient for implementation in standard clinical practice at the moment, inclusion of certain markers in predictive multifactorial models can improve their prognostic value. The future of the research in this field lies in the fine tuning of the clinical markers to be used, as well as identifying possible therapeutic techniques to prevent or reverse endothelial damage.
Collapse
Affiliation(s)
- Jakub Kornacki
- Department of Reproduction, Poznan University of Medical Sciences, 33 Polna Street, 60-535 Poznan, Poland; (J.K.); (E.W.-O.)
| | - Paweł Gutaj
- Department of Reproduction, Poznan University of Medical Sciences, 33 Polna Street, 60-535 Poznan, Poland; (J.K.); (E.W.-O.)
| | - Anastasia Kalantarova
- Medicine Program, Poznan University of Medical Sciences, 41 Jackowskiego Street, 60-512 Poznan, Poland;
| | - Rafał Sibiak
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Swiecickiego Street, 60-781 Poznan, Poland;
| | - Maurycy Jankowski
- Department of Anatomy, Poznan University of Medical Sciences, 6 Swiecickiego Street, 60-781 Poznan, Poland;
| | - Ewa Wender-Ozegowska
- Department of Reproduction, Poznan University of Medical Sciences, 33 Polna Street, 60-535 Poznan, Poland; (J.K.); (E.W.-O.)
| |
Collapse
|
13
|
Zhang J, Hua W, Zhao X, Yang F, Guo T, Zhang J, Zheng X, Liang W. Paeoniflorin alleviates endothelial dysfunction caused by overexpression of soluble fms-like tyrosine kinase 1 and soluble endoglin in preeclampsia via VEGFA upregulation. Biosci Biotechnol Biochem 2021; 85:814-823. [PMID: 33590855 DOI: 10.1093/bbb/zbaa106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 12/03/2020] [Indexed: 11/14/2022]
Abstract
This study assessed the protective effects of paeoniflorin against preeclampsia-related endothelial damage (ED). Human umbilical vein endothelial cells (HUVECs) isolated from healthy puerperae were identified by immunofluorescence assay. After paeoniflorin treatment, HUVECs were induced by soluble fms-like tyrosine kinase 1 (sFlt-1) and soluble endoglin (sEng) to establish ED. Cell viability, migration, invasion, tube formation, and apoptosis were assessed by (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) tetrazolium MTT assay, Scratch assay, Transwell assay, tube formation assay, and flow cytometry. VEGFA expression in HUVECs was analyzed by Western blot. HUVECs were successfully isolated and identified as Von Willebrand factor (vWF) positive. Individual treatment or cotreatment of sFlt-1 and sEng inhibited migration, invasion and tube formation, enhanced apoptosis, and decreased VEGFA expression in HUVECs. Paeoniflorin pretreatment partially reversed the effects delivered by cotreatment of sFlt-1 and sEng in HUVECs. Paeoniflorin alleviated preeclampsia-related ED caused by overexpression of sFlt-1 and sEng by upregulating VEGFA.
Collapse
Affiliation(s)
- Jin Zhang
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Wei Hua
- Reproductive Center, Xijing Hospital of Air Force Medical University, Xi'an City, Shan xi Province, China
| | - Xinyuan Zhao
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Fan Yang
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Ting Guo
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jianhua Zhang
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Xuerong Zheng
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Wanqi Liang
- The Second Department of Obstetrics, The Second Affiliated Hospital of Xi'an Medical University, Xi'an, China
| |
Collapse
|
14
|
Waker CA, Kaufman MR, Brown TL. Current State of Preeclampsia Mouse Models: Approaches, Relevance, and Standardization. Front Physiol 2021; 12:681632. [PMID: 34276401 PMCID: PMC8284253 DOI: 10.3389/fphys.2021.681632] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
Preeclampsia (PE) is a multisystemic, pregnancy-specific disorder and a leading cause of maternal and fetal death. PE is also associated with an increased risk for chronic morbidities later in life for mother and offspring. Abnormal placentation or placental function has been well-established as central to the genesis of PE; yet much remains to be determined about the factors involved in the development of this condition. Despite decades of investigation and many clinical trials, the only definitive treatment is parturition. To better understand the condition and identify potential targets preclinically, many approaches to simulate PE in mice have been developed and include mixed mouse strain crosses, genetic overexpression and knockout, exogenous agent administration, surgical manipulation, systemic adenoviral infection, and trophoblast-specific gene transfer. These models have been useful to investigate how biological perturbations identified in human PE are involved in the generation of PE-like symptoms and have improved the understanding of the molecular mechanisms underpinning the human condition. However, these approaches were characterized by a wide variety of physiological endpoints, which can make it difficult to compare effects across models and many of these approaches have aspects that lack physiological relevance to this human disorder and may interfere with therapeutic development. This report provides a comprehensive review of mouse models that exhibit PE-like symptoms and a proposed standardization of physiological characteristics for analysis in murine models of PE.
Collapse
Affiliation(s)
- Christopher A Waker
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Melissa R Kaufman
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Thomas L Brown
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| |
Collapse
|
15
|
Nuzzo AM, Giuffrida D, Moretti L, Re P, Grassi G, Menato G, Rolfo A. Placental and maternal sFlt1/PlGF expression in gestational diabetes mellitus. Sci Rep 2021; 11:2312. [PMID: 33504861 PMCID: PMC7840991 DOI: 10.1038/s41598-021-81785-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Gestational diabetes mellitus (GDM) and preeclampsia (PE) are both characterized by endothelial dysfunction and GDM women have higher incidence of PE. The placenta plays a key role in PE pathogenesis but its contribution to PE during GDM remains unclear. Herein, we compared placental and maternal blood anti-angiogenic soluble fms-like tyrosine kinase-1 (sFlt1) and pro-angiogenic Placental Growth Factor (PlGF) expressions in GDM and GDM-PE pregnancies compared to controls (CTRL) and PE cases. Electrochemiluminescence immunoassays showed a significantly higher maternal blood sFlt1/PlGF values in GDM-PE relative to CTRL and GDM pregnancies. We reported that placental PlGF gene expression was significantly decreased in GDM, PE and GDM-PE relative to CTRL. However, PlGF protein levels were significantly increased in GDM and GDM-PE relative to CTRL and PE placentae. Finally, sFlt1 gene expression was significantly increased in PE relative to CTRL, GDM and GDM-PE placentae. In contrast, sFlt1 protein expression was significantly decreased in GDM-PE relative to CTRL, GDM and PE placentae. Finally, higher sFlt1/PlGF ratio in GDM-PE maternal blood suggest that sFlt1 overproduction is related to PE onset also in GDM pregnancies even though characterized by a less severe endothelial dysfunction in terms of angiogenic biomarkers.
Collapse
Affiliation(s)
- Anna Maria Nuzzo
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Domenica Giuffrida
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Laura Moretti
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Paola Re
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Giorgio Grassi
- Department of Endocrinology, Diabetology, and Metabolism, Città della Salute e della Scienza University Hospital, Corso Bramante 88, 10126, Turin, Italy
| | - Guido Menato
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, Via Ventimiglia 3, 10126, Turin, Italy.
| |
Collapse
|
16
|
Yuan D, Yang Z, Chen Y, Li S, Tan B, Yu Q. Hypoxia-induced SPOP attenuates the mobility of trophoblast cells through inhibition of the PI3K/AKT/GSK3β pathway. Cell Biol Int 2021; 45:599-611. [PMID: 33200474 DOI: 10.1002/cbin.11501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/20/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022]
Abstract
Placental hypoxia has been implicated in pregnancy pathologies such as pre-eclampsia and intrauterine growth restriction. However, the underlying mechanism by which the trophoblasts respond to hypoxia remains unclear. Speckle-type POZ protein (SPOP), an E3 ubiquitin ligase adapter, was previously reported to play important roles in various physiological and pathological processes. This study aims to investigate the expression and biological functions of SPOP after exposure to cobalt chloride (CoCl2 )-mimicked hypoxia conditions using human trophoblast-derived choriocarcinoma cell lines and extravillous cytotrophoblast. These data showed that SPOP protein was directly induced by CoCl2 -mimicked hypoxia and regulated by HIF-1α at the posttranscription level. CoCl2 treatment could dramatically influence the localization of SPOP in trophoblasts, especially the accumulation of SPOP into the nucleus. In addition, both CoCl2 -mimicked hypoxia and induction of endogenous SPOP expression by lentivirus transfection attenuated the migration and invasion abilities of trophoblasts. Furthermore, we demonstrated that SPOP was involved in CoCl2 -induced the inhibition of the PI3K/AKT/GSK3β pathway in placental trophoblasts. Taken together, these data indicate that accumulation of HIF-1α augments the expression of SPOP in trophoblasts, which impairs trophoblastic mobility by targeting the PI3K/AKT/GSK3β pathway. This potentially leads to insufficient uterine spiral artery remodeling and suboptimal placental perfusion, and thus the development of pregnancy-related complication.
Collapse
Affiliation(s)
- Dong Yuan
- Department of Gynecology, Chongqing Medical University Affiliated Second Hospital, Chongqing, China
| | - Zhu Yang
- Department of Gynecology, Chongqing Medical University Affiliated Second Hospital, Chongqing, China.,Molecular Medical Laboratory, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Yiyu Chen
- Department of Clinical Laboratory, Chongqing Medical University Affiliated Stomatological Hospital, Chongqing, China
| | - Siyuan Li
- Department of Laboratory Medicine, Chongqing Medical University Affiliated Children's Hospital, Chongqing, China
| | - Benxu Tan
- Department of Oncology, Chongqing Medical University Affiliated First Hospital, Chongqing, China
| | - Qiubo Yu
- Molecular Medical Laboratory, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Kitjaras T, Phupong V. Maternal serum split and hairy-related protein 1 in preeclampsia versus normal pregnancy. J Matern Fetal Neonatal Med 2020; 35:4179-4183. [PMID: 33207988 DOI: 10.1080/14767058.2020.1849090] [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: 10/22/2022]
Abstract
OBJECTIVE The primary objective was to compare maternal serum split and hairy related protein-1 (SHARP1) in preeclamptic women versus normal pregnant women. The secondary objectives were to compare maternal and neonatal outcomes between preeclampsia and normal pregnant women. METHODS This cross-sectional analytic study included 75 preeclamptic pregnant women and 75 normal pregnant women at Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital between March 2018 and May 2019. Blood was collected into nonheparinized tubes. Blood samples were centrifuged and stored at -80 °C until assayed. Maternal serum SHARP1 levels were measured by enzyme-linked immunosorbent assays. Maternal and neonatal outcomes were recorded. RESULTS Median serum SHARP1 level in preeclampsia was significantly higher than in normal pregnancy (5.2 vs. 3.7 ng/mL, p = .007). Preeclamptic women had a higher rate of cesarean section than normal pregnant women (68% vs. 32%, p < .001). Preeclamptic women had lower neonatal birth weight than normal pregnant women (2,546.1 ± 766.3 vs 2,884.5 ± 479.9, p = .001). Neonatal complications were higher in preeclamptic women than normal pregnant women (40% vs. 17.3%, p = .002). CONCLUSIONS Serum SHARP1 in preeclampsia was significantly higher than in normal pregnancy.
Collapse
Affiliation(s)
- Thanisa Kitjaras
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Vorapong Phupong
- Department of Obstetrics and Gynecology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
18
|
Stenhouse C, Hogg CO, Ashworth CJ. Novel relationships between porcine fetal size, sex, and endometrial angiogenesis†. Biol Reprod 2020; 101:112-125. [PMID: 31093645 DOI: 10.1093/biolre/ioz068] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 01/17/2019] [Accepted: 04/22/2019] [Indexed: 11/13/2022] Open
Abstract
It is hypothesized that growth restriction occurs due to inadequate vascularization of the feto-maternal interface. Evidence exists for sexual dimorphism in placental function although associations between fetal sex and the endometrium remain poorly investigated. This study investigated the relationship between porcine fetal size, sex and endometrial angiogenesis at multiple gestational days (GD). Endometrial samples supplying the lightest and closest to mean litter weight (CTMLW), male and female Large White X Landrace conceptuses or fetuses were obtained at GD18, 30, 45, 60, and 90 (n = 5-9 litters/GD). Immunohistochemistry for CD31 revealed a greater number of blood vessels in endometrium supplying females compared to those supplying males at GD45. Endometrial samples supplying the lightest fetuses had fewer blood vessels (GD60) and uterine glands (GD90) compared to those supplying the CTMLW fetuses. Quantitative PCR revealed decreased CD31 (GD60), HPSE and VEGFA (GD90) expression, alongside increased HIF1A (GD45) expression in endometrial samples supplying the lightest compared to the CTMLW fetuses. At GD30, PTGFR, CD31, and VEGFA mRNA expression was increased in samples supplying female fetuses compared to those supplying male fetuses. Intriguingly, decreased expression of ACP5, CD31, HIF1A, and VEGFA mRNAs was observed at GD60 in endometrial samples supplying female fetuses compared to those supplying their male littermates. Endothelial cell branching assays demonstrated impaired endothelial cell branching in response to conditioned media from endometrial samples supplying the lightest and female fetuses compared with the CTMLW and male fetuses, respectively. This study has highlighted that endometrial tissues supplying the lightest and female fetuses have impaired angiogenesis when compared with the CTMLW and female fetuses respectively. Importantly, the relationship between fetal size, sex and endometrial vascularity is dynamic and dependent upon the GD investigated.
Collapse
Affiliation(s)
- Claire Stenhouse
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian, UK
| | - Charis O Hogg
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian, UK
| | - Cheryl J Ashworth
- Developmental Biology Division, The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Midlothian, UK
| |
Collapse
|
19
|
Fan J, Yu Y, Han X, He H, Luo Y, Yu S, Cui Y, Xu G, Wang L, Pan Y. The expression of hypoxia-inducible factor-1 alpha in primary reproductive organs of the female yak (Bos grunniens) at different reproductive stages. Reprod Domest Anim 2020; 55:1371-1382. [PMID: 32706432 DOI: 10.1111/rda.13783] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/19/2020] [Indexed: 12/12/2022]
Abstract
The yak (Bos grunniens) is the most important livestock animal in high-altitude regions owing to its prominent adaptability to cold conditions, nutritional deficiencies and hypoxia. The reproductive organs exhibit different histological appearances and physiological processes at different reproductive stages. Hypoxia-inducible factor-1 alpha (HIF-1α) is the regulatory subunit of HIF-1 that crucially regulates the response to hypoxia in mammalian organisms. The goal of our study was to investigate the expression and distribution of HIF-1α in the primary yak reproductive organs at different reproductive stages. Samples of the ovary, oviduct and uterus of 15 adult female yaks were collected and used in the experiment. The expression and localization of HIF-1α proteins and mRNA were investigated using quantitative real-time polymerase chain reaction (qRT-PCR), Western blot (WB) and immunohistochemistry (IHC). The results indicated that the expression of HIF-1α protein in the ovary was higher during the luteal phase than during the follicular phase and gestation period (p < .05). In the oviduct, HIF-1α protein was also more highly expressed during the luteal phase than during the follicular phase and gestation period (p < .01). However, in the uterus, the HIF-1α protein had stronger expression during the gestation period than during the follicular phase (p < .01) and luteal phase (p < .05). The expression of HIF-1α mRNA was similar to that of its protein. Immunohistochemical analysis revealed intense immunostaining of HIF-1α proteins in the follicular granulosa cells, granular luteal cells, villous epithelial cells of the oviduct, endometrial glandular epithelium and luminal epithelium, foetal villous trophoblast, and epithelia of caruncular crypts. This study showed that the expression of HIF-1α in the ovary, oviduct and uterus varies according to the stage of the reproductive cycle. This implies that HIF-1α plays an important role in regulating the stage-specific physiological function of yak reproductive organs under hypoxic environments.
Collapse
Affiliation(s)
- Jiangfeng Fan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yiteng Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xiaohong Han
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Honghong He
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yuzhu Luo
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Sijiu Yu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yan Cui
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, China
| | - Gengquan Xu
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, China
| | - Libin Wang
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, China
| | - Yangyang Pan
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, China
| |
Collapse
|
20
|
HIF-1α immunohistochemical expression in decidual cells, villous and extravillous trophoblast in placentas from pregnancies complicated with preeclampsia. Pregnancy Hypertens 2020; 21:176-178. [PMID: 32563172 DOI: 10.1016/j.preghy.2020.06.003] [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: 09/20/2019] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 11/23/2022]
Abstract
Etiology of preeclampsia (PE) remains unknown despite it being the leading cause of maternal and neonatal morbidity and mortality worldwide. PE placentas show signs of chronic oxidative stress, therefore we set to investigate the HIF-1α immunohistochemical expression in PE placentas. For the first time presented study reported on significantly higher HIF-1α immunohistochemical expression in all of the placental segments; villous trophoblast (VTB), decidual cells (DC) and extravillous trophoblast (EVTB) in the PE placentas compared to placentas from normal pregnancies. The further studies on the subject are necessary in order to understand the mechanisms behind elevated HIF-1α immunohistochemical levels in PE.
Collapse
|
21
|
Frazier S, McBride MW, Mulvana H, Graham D. From animal models to patients: the role of placental microRNAs, miR-210, miR-126, and miR-148a/152 in preeclampsia. Clin Sci (Lond) 2020; 134:1001-1025. [PMID: 32337535 PMCID: PMC7239341 DOI: 10.1042/cs20200023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/23/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022]
Abstract
Placental microRNAs (miRNAs) regulate the placental transcriptome and play a pathological role in preeclampsia (PE), a hypertensive disorder of pregnancy. Three PE rodent model studies explored the role of placental miRNAs, miR-210, miR-126, and miR-148/152 respectively, by examining expression of the miRNAs, their inducers, and potential gene targets. This review evaluates the role of miR-210, miR-126, and miR-148/152 in PE by comparing findings from the three rodent model studies with in vitro studies, other animal models, and preeclamptic patients to provide comprehensive insight into genetic components and pathological processes in the placenta contributing to PE. The majority of studies demonstrate miR-210 is upregulated in PE in part driven by HIF-1α and NF-κBp50, stimulated by hypoxia and/or immune-mediated processes. Elevated miR-210 may contribute to PE via inhibiting anti-inflammatory Th2-cytokines. Studies report an up- and downregulation of miR-126, arguably reflecting differences in expression between cell types and its multifunctional capacity. MiR-126 may play a pro-angiogenic role by mediating the PI3K-Akt pathway. Most studies report miR-148/152 family members are upregulated in PE. Evidence suggests they may inhibit DNA methylation of genes involved in metabolic and inflammatory pathways. Given the genetic heterogeneity of PE, it is unlikely that a single placental miRNA is a suitable therapeutic target for all patients. Investigating miRNAs in PE subtypes in patients and animal models may represent a more appropriate approach going forward. Developing methods for targeting placental miRNAs and specific placental cell types remains crucial for research seeking to target placental miRNAs as a novel treatment for PE.
Collapse
Affiliation(s)
- Sonya Frazier
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Martin W. McBride
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Helen Mulvana
- Biomedical Engineering, University of Strathclyde, Glasgow, U.K
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| |
Collapse
|
22
|
Zhang Z, Huang C, Wang P, Gao J, Liu X, Li Y, Yan S, Shi Y. HIF‑1α affects trophoblastic apoptosis involved in the onset of preeclampsia by regulating FOXO3a under hypoxic conditions. Mol Med Rep 2020; 21:2484-2492. [PMID: 32323858 PMCID: PMC7185296 DOI: 10.3892/mmr.2020.11050] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 02/07/2020] [Indexed: 12/25/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy-specific syndrome that has severe implications on perinatal mortality and morbidity. Excessive apoptosis of trophoblasts induced by hypoxia may be associated with the development of PE, but the exact pathogenesis is unknown. Forkhead box O transcription factor 3a (FOXO3a) is activated under hypoxic conditions. Furthermore, hypoxia-inducible factor-1α (HIF-1α) is sensitive to variations in partial oxygen pressure. Thus, the aims of the present study were to investigate the expression levels of HIF-1α and FOXO3a in placental samples of early onset severe PE, and their effect on trophoblastic apoptosis under hypoxic conditions. Cobalt chloride was used to establish the hypoxic model. The present study examined the expression levels of HIF-1α and FOXO3a in the placental tissues and HTR8/SVneo cells under hypoxic conditions. It was found that HIF-1α and FOXO3a were highly expressed in placental tissues of patients with PE and in HTR8/SVneo cells under hypoxic conditions. Furthermore, knockdown of FOXO3a using a specific small interfering RNA (siRNA) decreased apoptosis in HTR8/SVneo cells. Moreover, it was found that after knockdown of HIF-1α using siRNA, FOXO3a expression and the apoptotic rate were reduced in HTR8/SVneo cells. Therefore, the present results indicated that the elevated expression of HIF-1α increased trophoblastic apoptosis by regulating FOXO3a, which may be involved in the pathogenesis of PE.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chenxi Huang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ping Wang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Junjun Gao
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xin Liu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yingying Li
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shujun Yan
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ying Shi
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
23
|
Song W, Chang WL, Shan D, Gu Y, Gao L, Liang S, Guo H, Yu J, Liu X. Intermittent Hypoxia Impairs Trophoblast Cell Viability by Triggering the Endoplasmic Reticulum Stress Pathway. Reprod Sci 2020; 27:477-487. [PMID: 32016801 DOI: 10.1007/s43032-019-00039-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 07/31/2019] [Indexed: 01/24/2023]
Abstract
Intermittent hypoxia (IH) is a prominent characteristic of many clinical complications such as obstructive sleep apnea syndrome (OSAS). OSAS is related to a higher incidence of adverse pregnancy outcomes, and IH has been suggested as the preliminary physiological etiology. However, further studies remain to be performed on the underlying cellular and molecular pathogenic mechanisms of OSAS-related IH on adverse pregnancy outcomes. Here, we used a trophoblast cell line (HTR8/SVneo), primary extravillous trophoblast cells (EVTs), and a normal-term placenta villi explant culture model in vitro in this research. The effects and possible molecular mechanisms of IH on trophoblast motility, cell cycle progression, and apoptosis were investigated. IH reduced HTR8/SVneo cell and EVT motility significantly, which could be partially attributed to the reduced secretion of matrix metalloproteinase 2. IH treatment blocked HTR8/SVneo cell proliferation significantly by modulating the expression of D-type Cyclins. IH also induced significant trophoblast cell apoptosis. Moreover, our study supports the premise that IH attenuates trophoblast cell motility and proliferation and induces excessive trophoblast cell apoptosis by specifically triggering the endoplasmic reticulum (ER) stress signaling pathway. Briefly, differing from the mechanism of trophoblast motility and proliferation inhibition, and apoptosis induction by hypoxia, IH is apt to weaken trophoblast viability mainly by activating the ER stress signaling pathway with a time-dependent pattern, which is further implicated in OSAS-associated adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Wei Song
- Department of Beijing Obstetric and Gyneocology Hospital, Capital Medical University, Beijing, 100026, China
| | - Wen-Lin Chang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Department of Obstetrics, the People' Hospital of Longhua, Shenzhen, 518109, China
| | - Dan Shan
- Department of Beijing Obstetric and Gyneocology Hospital, Capital Medical University, Beijing, 100026, China
| | - Yanli Gu
- Central Laboratory, the People' Hospital of Longhua, Shenzhen, 518109, China
| | - Lei Gao
- Department of Beijing Obstetric and Gyneocology Hospital, Capital Medical University, Beijing, 100026, China
| | - Shengnan Liang
- Department of Beijing Obstetric and Gyneocology Hospital, Capital Medical University, Beijing, 100026, China
| | - Huan Guo
- Department of Urology, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, 518000, China
| | - Jing Yu
- Department of Laboratory Medicine, Peking University Shenzhen Hospital, 1120 Lianhua Road, Futian District, Shenzhen, 518036, Guangdong, China.
| | - Xiaowei Liu
- Department of Beijing Obstetric and Gyneocology Hospital, Capital Medical University, Beijing, 100026, China.
| |
Collapse
|
24
|
Owaki Y, Watanabe K, Iwasaki A, Saitou T, Matsushita H, Wakatsuki A. Placental hypoplasia and maternal organic vascular disorder in pregnant women with gestational hypertension and preeclampsia. J Matern Fetal Neonatal Med 2019; 34:353-359. [PMID: 30983440 DOI: 10.1080/14767058.2019.1608175] [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: 10/27/2022]
Abstract
Objective: This study aimed to investigate the etiology and pathology of preeclampsia (PE), a two-stage disorder involving uteroplacental dysfunction resulting from abnormal implantation and placentation, and gestational hypertension (GH), for which maternal organic vascular disorder is often an underlying factor.Methods: We assessed concentrations of oxygen free radicals (d-ROMs), maternal angiogenic factor (PlGF), and antiangiogenic factor (sFlt-1), placental hypoxic changes, oxidative DNA damage, and maternal organic vascular disorders in 23 women with PE (PE group), 13 with GH (GH group), and 16 with uncomplicated pregnancies (normal group). Intima-media thickness (IMT) of the carotid artery was assessed as a proxy for maternal organ vascular disorder. Immunohistochemical analysis was performed to measure the proportion of placental trophoblast cell nuclei staining positive for hypoxia-inducible factor-1α (HIF-1α), which reflects hypoxic changes, and 8-hydroxy-2'-deoxyguanosine (8-OHdG), which reflects oxidative DNA damage.Results: Maternal serum d-ROM concentrations were significantly increased in both GH and PE groups relative to the normal group. Maternal serum d-ROM concentrations were significantly increased in both GH and PE groups relative to the normal group. Maternal serum sFlt-1 concentrations, ratio of sFlt-1/PlGF, and proportions of HIF-1α-positive nuclei and 8-OHdG-positive nuclei were significantly higher in the PE group compared to GH and normal groups. IMT was significantly greater in GH and PE groups compared to the normal group, and was higher in the GH group compared to the PE group.Conclusions: Our findings suggest that placental hypoxic changes and oxidative DNA damage are severe in patients with PE and accompanied by an increase in antiangiogenic factors. Moreover, maternal organ vascular disorder was more severe in patients with GH compared to those with PE, as assessed by IMT.Key message: PE is a two-stage disorder that involves uteroplacental dysfunction, and organic vascular disorder underlies GH.
Collapse
Affiliation(s)
- Yuki Owaki
- Department of Obstetrics and Gynecology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Kazushi Watanabe
- Department of Obstetrics and Gynecology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Ai Iwasaki
- Department of Obstetrics and Gynecology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Takuya Saitou
- Department of Obstetrics and Gynecology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hiroshi Matsushita
- Department of Obstetrics and Gynecology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Akihiko Wakatsuki
- Department of Obstetrics and Gynecology, Aichi Medical University School of Medicine, Nagakute, Japan
| |
Collapse
|
25
|
Farrell A, Alahari S, Ermini L, Tagliaferro A, Litvack M, Post M, Caniggia I. Faulty oxygen sensing disrupts angiomotin function in trophoblast cell migration and predisposes to preeclampsia. JCI Insight 2019; 4:127009. [PMID: 30996134 DOI: 10.1172/jci.insight.127009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 03/14/2019] [Indexed: 12/17/2022] Open
Abstract
Human placenta development and a successful pregnancy is incumbent upon precise oxygen-dependent control of trophoblast migration/invasion. Persistent low oxygen leading to failed trophoblast invasion promotes inadequate spiral artery remodeling, a characteristic of preeclampsia. Angiomotin (AMOT) is a multifaceted scaffolding protein involved in cell polarity and migration, yet its upstream regulation and significance in the human placenta remain unknown. Herein, we show that AMOT is primarily expressed in migratory extravillous trophoblast cells (EVTs) of the intermediate and distal anchoring column. Its expression increases after 10 weeks of gestation when oxygen tension rises and EVT migration/invasion peaks. Time-lapse imaging confirmed that the AMOT 80-kDa isoform promotes migration of trophoblastic JEG3 and HTR-8/SVneo cells. In preeclampsia, however, AMOT expression is decreased and its localization to migratory fetomaternal interface EVTs is disrupted. We demonstrate that Jumonji C domain-containing protein 6 (JMJD6), an oxygen sensor, positively regulates AMOT via oxygen-dependent lysyl hydroxylation. Furthermore, in vitro and ex vivo studies show that transforming growth factor-β (TGF-β) regulates AMOT expression, its interaction with polarity protein PAR6, and its subcellular redistribution from tight junctions to cytoskeleton. Our data reveal an oxygen- and TGF-β-driven migratory function for AMOT in the human placenta, and implicate its deficiency in impaired trophoblast migration that plagues preeclampsia.
Collapse
Affiliation(s)
- Abby Farrell
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences, and
| | - Sruthi Alahari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Leonardo Ermini
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Andrea Tagliaferro
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Michael Litvack
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Martin Post
- Institute of Medical Sciences, and.,Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Institute of Medical Sciences, and.,Department of Physiology, University of Toronto, Toronto, Ontario, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Abstract
Complications of pregnancy remain key drivers of morbidity and mortality, affecting the health of both the mother and her offspring in the short and long term. There is lack of detailed understanding of the pathways involved in the pathology and pathogenesis of compromised pregnancy, as well as a shortfall of effective prognostic, diagnostic and treatment options. In many complications of pregnancy, such as in preeclampsia, there is an increase in uteroplacental vascular resistance. However, the cause and effect relationship between placental dysfunction and adverse outcomes in the mother and her offspring remains uncertain. In this review, we aim to highlight the value of gestational hypoxia-induced complications of pregnancy in elucidating underlying molecular pathways and in assessing candidate therapeutic options for these complex disorders. Chronic maternal hypoxia not only mimics the placental pathology associated with obstetric syndromes like gestational hypertension at morphological, molecular and functional levels, but also recapitulates key symptoms that occur as maternal and fetal clinical manifestations of these pregnancy disorders. We propose that gestational hypoxia provides a useful model to study the inter-relationship between placental dysfunction and adverse outcomes in the mother and her offspring in a wide array of examples of complicated pregnancy, such as in preeclampsia.
Collapse
|
27
|
Trophoblast-Specific Expression of Hif-1α Results in Preeclampsia-Like Symptoms and Fetal Growth Restriction. Sci Rep 2019; 9:2742. [PMID: 30808910 PMCID: PMC6391498 DOI: 10.1038/s41598-019-39426-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 12/17/2018] [Indexed: 01/02/2023] Open
Abstract
The placenta is an essential organ that is formed during pregnancy and its proper development is critical for embryonic survival. While several animal models have been shown to exhibit some of the pathological effects present in human preeclampsia, these models often do not represent the physiological aspects that have been identified. Hypoxia-inducible factor 1 alpha (Hif-1α) is a necessary component of the cellular oxygen-sensing machinery and has been implicated as a major regulator of trophoblast differentiation. Elevated levels of Hif-1α in the human placenta have been linked to the development of pregnancy-associated disorders, such as preeclampsia and fetal growth restriction. As oxygen regulation is a critical determinant for placentogenesis, we determined the effects of constitutively active Hif-1α, specifically in trophoblasts, on mouse placental development in vivo. Our research indicates that prolonged expression of trophoblast-specific Hif-1α leads to a significant decrease in fetal birth weight. In addition, we noted significant physiological alterations in placental differentiation that included reduced branching morphogenesis, alterations in maternal and fetal blood spaces, and failure to remodel the maternal spiral arteries. These placental alterations resulted in subsequent maternal hypertension with parturitional resolution and maternal kidney glomeruloendotheliosis with accompanying proteinuria, classic hallmarks of preeclampsia. Our findings identify Hif-1α as a critical molecular mediator of placental development and indicate that prolonged expression of Hif-1α, explicitly in placental trophoblasts causes maternal pathology and establishes a mouse model that significantly recapitulates the physiological and pathophysiological characteristics of preeclampsia with fetal growth restriction.
Collapse
|
28
|
Glycosylation Profile of the Transferrin Receptor in Gestational Iron Deficiency and Early-Onset Severe Preeclampsia. J Pregnancy 2019; 2019:9514546. [PMID: 30854239 PMCID: PMC6378037 DOI: 10.1155/2019/9514546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 12/01/2018] [Accepted: 01/15/2019] [Indexed: 12/14/2022] Open
Abstract
Objective To examine the expression of hypoxia-inducible factor-1α (HIF-1α), TfR1, and TfR1-attached terminal monosaccharides in placentas of women with IDAP and severe preeclampsia. Methods TfR1 and HIF-1α were detected by western blot. Immunoadsorption of TfR1 was performed to characterize the terminal monosaccharides by specific lectin binding. Results There was no difference in the expression of TfR1 and HIF-1α between groups. Lectin blot analysis pointed out an overexpression of galactose β1-4 N-acetylglucosamine (Gal-GlcNAc) and mannose in severe preeclampsia. Conclusion The increase in Gal-GlcNAc may be due to the increased presence of antennary structures and the mannose glycans of TfR1 may indicate the presence of misfolded or incomplete proteins. These findings may be associated with the low expression of placental TfR1 in women with preeclampsia.
Collapse
|
29
|
Chaudhary P, Babu GS, Sobti RC, Gupta SK. HGF regulate HTR-8/SVneo trophoblastic cells migration/invasion under hypoxic conditions through increased HIF-1α expression via MAPK and PI3K pathways. J Cell Commun Signal 2019; 13:503-521. [PMID: 30684191 DOI: 10.1007/s12079-019-00505-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 01/17/2019] [Indexed: 12/22/2022] Open
Abstract
Hepatocyte growth factor (HGF) is reported to be down-regulated in pregnancy complications like intrauterine growth retardation and preeclampsia, which are associated with abnormal trophoblast migration/invasion. In this study, role of HGF and associated signaling pathways has been investigated in HTR-8/SVneo trophoblastic cells migration/invasion under normoxia (20% O2) and hypoxia (2% O2). HTR-8/SVneo cells exposed to hypoxia showed increase in migration and invasion as compared to cells incubated under normoxic conditions. The migration/invasion under both normoxic and hypoxic conditions was further enhanced after treatment with HGF. Subsequent to treatment with HGF, a significant increase in expression of MMP2 & MMP3 under normoxia and MMP1 & MMP9 under hypoxia was observed. Treatment of HTR-8/SVneo cells with HGF under hypoxia also led to decrease in TIMP1. Treatment of the cells with HGF led to activation of mitogen activated protein kinases (MAPK) and phosphatidylinositol 3-kinase (PI3K) signaling pathways. Inhibition of MAPK by U0126 and PI3K by LY294002 led to concomitant decrease in the HGF-mediated migration/invasion of HTR-8/SVneo cells. HGF treatment under hypoxia also led to a significant increase in hypoxia inducible factor (HIF-1α) expression. Additionally, inhibition of HIF-1α by siRNA led to decrease in HGF-mediated migration of HTR-8/SVneo cells under hypoxic conditions. Inhibition of HGF activated MAPK and PI3K signaling led to reduction in HIF-1α expression under hypoxia. In conclusion, HGF facilitates HTR-8/SVneo cell migration/invasion by activation of MAPK/PI3K signaling pathways and increased expression of MMPs. HIF-1α has a role in HGF-mediated increase in migration under hypoxic conditions.
Collapse
Affiliation(s)
- Piyush Chaudhary
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India.,Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Lucknow, 226 025, India
| | - Gosipatala Sunil Babu
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Lucknow, 226 025, India
| | - Ranbir Chander Sobti
- Department of Biotechnology, Panjab University, Sector-14, Chandigarh, 160 014, India
| | - Satish Kumar Gupta
- Reproductive Cell Biology Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, 110 067, India.
| |
Collapse
|
30
|
Alahari S, Garcia J, Post M, Caniggia I. The von Hippel Lindau tumour suppressor gene is a novel target of E2F4-mediated transcriptional repression in preeclampsia. Biochim Biophys Acta Mol Basis Dis 2018; 1864:3298-3308. [DOI: 10.1016/j.bbadis.2018.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/20/2018] [Accepted: 07/16/2018] [Indexed: 12/31/2022]
|
31
|
Soares MJ, Iqbal K, Kozai K. Hypoxia and Placental Development. Birth Defects Res 2018; 109:1309-1329. [PMID: 29105383 DOI: 10.1002/bdr2.1135] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 09/04/2017] [Indexed: 12/17/2022]
Abstract
Hemochorial placentation is orchestrated through highly regulated temporal and spatial decisions governing the fate of trophoblast stem/progenitor cells. Trophoblast cell acquisition of specializations facilitating invasion and uterine spiral artery remodeling is a labile process, sensitive to the environment, and represents a process that is vulnerable to dysmorphogenesis in pathologic states. Hypoxia is a signal guiding placental development, and molecular mechanisms directing cellular adaptations to low oxygen tension are integral to trophoblast cell differentiation and placentation. Hypoxia can also be used as an experimental tool to investigate regulatory processes controlling hemochorial placentation. These developmental processes are conserved in mouse, rat, and human placentation. Consequently, elements of these developmental events can be modeled and hypotheses tested in trophoblast stem cells and in genetically manipulated rodents. Hypoxia is also a consequence of a failed placenta, yielding pathologies that can adversely affect maternal adjustments to pregnancy, fetal health, and susceptibility to adult disease. The capacity of the placenta for adaptation to environmental challenges highlights the importance of its plasticity in safeguarding a healthy pregnancy. Birth Defects Research 109:1309-1329, 2017.© 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Michael J Soares
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas.,Department of Pediatrics, University of Kansas Medical Center, Kansas City, Kansas.,Fetal Health Research, Children's Research Institute, Children's Mercy, Kansas City, Missouri
| | - Khursheed Iqbal
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Keisuke Kozai
- Institute for Reproduction and Perinatal Research, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
32
|
Monteiro LJ, Cubillos S, Sanchez M, Acuña-Gallardo S, Venegas P, Herrera V, Lam EWF, Varas-Godoy M, Illanes SE. Reduced FOXM1 Expression Limits Trophoblast Migration and Angiogenesis and Is Associated With Preeclampsia. Reprod Sci 2018; 26:580-590. [PMID: 29848205 DOI: 10.1177/1933719118778798] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Trophoblast cells are often compared to highly invasive carcinoma cells due to their capacity to proliferate in hypoxic conditions and to exhibit analogous vascular, proliferative, migratory, and invasive capacities. Thus, genes that are important for tumorigenesis, such as forkhead box M1 ( FOXM1) may also be involved in processes of trophoblast invasion. Indeed, we found Foxm1 protein and messenger RNA (mRNA) levels decreased as gestational age increased in rat's placentae. Accordingly, when mimicking early placental events in vitro, protein and mRNA expression of FOXM1 increased from 21% to 8% O2, reaching its highest expression at 3% oxygen tension, which reflects early implantation environment, and dropping to very low levels at 1% O2. Remarkably, FOXM1 silencing in JEG-3 cells was able to significantly decrease migration by 27.9%, in comparison with those cells transfected with control siRNA. Moreover, angiogenesis was compromised when conditioned media (CM) from FOXM1-siRNA -JEG-3 (3% O2) was added to human umbilical vein endothelial cells (HUVEC) cells; however, when CM of JEG-3 cells overexpressing FOXM1 at 1% O2 was added, the ability of HUVEC to form tubule networks was restored. Additionally, quantitative real-time polymerase chain reaction (PCR) assays of FOXM1 knockdown and overexpression experiments in JEG-3 cells revealed that the depletion of FOXM1 at 3% O2 and overexpression of FOXM1 at 1% O2 led to downregulation and upregulation of vascular endothelial growth factor transcriptional (VEGF) levels, respectively. Conversely, we also observed deregulation of FOXM1 in placentae derived from pregnancies complicated by preeclampsia (PE). Therefore, we demonstrate that FOXM1 may be a new regulatory protein of early placentation processes and that under chronic hypoxic conditions (1% O2) and in patients with severe PE, its levels decrease.
Collapse
Affiliation(s)
- Lara J Monteiro
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Sofia Cubillos
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Marianela Sanchez
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Stephanie Acuña-Gallardo
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Pía Venegas
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,2 Department of Maternal-Foetal Medicine, Clínica Dávila, Santiago, Chile
| | - Valentina Herrera
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Eric W-F Lam
- 3 Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College London, Hammersmith Hospital Campus, London, United Kingdom
| | - Manuel Varas-Godoy
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile
| | - Sebastián E Illanes
- 1 Laboratory of Reproductive Biology, Centre for Biomedical Research, Faculty of Medicine, Universidad de los Andes, Santiago, Chile.,2 Department of Maternal-Foetal Medicine, Clínica Dávila, Santiago, Chile
| |
Collapse
|
33
|
Ali LE, Salih MM, Elhassan EM, Mohmmed AA, Adam I. Placental growth factor, vascular endothelial growth factor, and hypoxia-inducible factor-1α in the placentas of women with pre-eclampsia. J Matern Fetal Neonatal Med 2018; 32:2628-2632. [PMID: 29455633 DOI: 10.1080/14767058.2018.1443066] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Although the exact mechanism of pre-eclampsia - high blood pressure and proteinuria after 20 gestational weeks - is not yet fully understood, placental growth factor (PLGF), vascular endothelial growth factor (VEGF), and hypoxia-inducible factor (HIF) are known to play important roles in vascularization and in the pathology of pre-eclampsia. METHODS PLGF, VEGF, and HIF-1α were evaluated by immunohistochemistry in the placentas of Sudanese women with mild or severe pre-eclampsia, and in normal controls. RESULTS Sixty-two women had severe pre-eclampsia, 102 had mild pre-eclampsia and 101 women served as healthy controls. Immunohistochemical staining of PLGF was significantly lower in placentas of women with severe pre-eclampsia (16%) compared with those with mild pre-eclampsia (8.8%) and placentas of normotensive women (40.6%; p < .001). Significantly more of the pre-eclamptic placentas expressed VEGF: in 32%, 17.6%, and 14.9% (p = .020) of the placentas of women with severe or mild pre-eclampsia and in controls, respectively. Significantly more of the pre-eclamptic placentas expressed HIF-1α: in 15%, 10.8%, and 5.0% of the placentas of women with severe or mild pre-eclampsia, and in controls, respectively (p = .044). CONCLUSION The current study showed that PLGF, VEGF, and HIF-1α are involved in the pathophysiology of pre-eclampsia.
Collapse
Affiliation(s)
- Lobna E Ali
- a Department of Histopathology and Cytology, Ahlia College of Medical Laboratory Sciences , Port Sudan , Sudan
| | - Magdi M Salih
- b Faculty of Medical Laboratory Sciences , University of Khartoum , Khartoum , Sudan
| | | | | | - Ishag Adam
- b Faculty of Medical Laboratory Sciences , University of Khartoum , Khartoum , Sudan
| |
Collapse
|
34
|
Sallais J, Alahari S, Tagliaferro A, Bhattacharjee J, Post M, Caniggia I. Factor inhibiting HIF1-A novel target of SUMOylation in the human placenta. Oncotarget 2017; 8:114002-114018. [PMID: 29371964 PMCID: PMC5768381 DOI: 10.18632/oncotarget.23113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 11/15/2017] [Indexed: 01/07/2023] Open
Abstract
Adaptations to changes in oxygen are critical to ensure proper placental development, and impairments in oxygen sensing mechanisms characterize placental pathologies such as preeclampsia. In this study, we examined the involvement of SUMOylation, a reversible posttranslational modification, in the regulation of the asparaginyl hydroxylase Factor Inhibiting Hypoxia Inducible Factor 1 (FIH1) in the human placenta in development and in disease status. FIH1 protein abundance and spatial distribution in the developing placenta directly correlated with oxygen tension in vivo. Immunofluorescence analysis showed that early on FIH1 primarily localized to nuclei of cytotrophoblast cells, while after 10 weeks of gestation it was present in nuclei and cytoplasm of both cytotrophoblast and syncytiotrophoblast cells. Exposure of choriocarcinoma JEG-3 cells to hypoxia induced FIH1 SUMOylation by promoting its association to SUMO2/3. Transfection of JEG-3 cells with FIH1 constructs containing SUMO-mutated sites revealed that SUMOylation of FIH1 by SUMO2/3 targeted it for proteasomal degradation, particularly in hypoxia. SUMOylation of FIH1 directly impacted on HIF1A activity as determined by HIF-responsive luciferase assay. Co-immunoprecipitation analyses revealed enhanced FIH1-SUMO2/3 associations early in development, when FIH1 levels are low, while deSUMOylation of FIH1 by SENP3 increased later in gestation, when FIH1 levels are rising. In preeclampsia, decreased FIH1 protein expression associated with impaired deSUMOylation by SENP3 and increased association with the ubiquitin ligase RNF4. We propose a novel mode of regulation of FIH1 stability by dynamic SUMOylation and deSUMOylation in the human placenta in response to changing oxygen tension, thereby mediating HIF1A transcriptional activity in physiological and pathological conditions.
Collapse
Affiliation(s)
- Julien Sallais
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Ontario, Canada
| | - Sruthi Alahari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada
| | - Andrea Tagliaferro
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Jayonta Bhattacharjee
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Martin Post
- Department of Obstetrics and Gynaecology, University of Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada.,Program in Physiology and Experimental Medicine, Peter Gilgan Centre for Research and Learning, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Obstetrics and Gynaecology, University of Toronto, Ontario, Canada.,Department of Physiology, University of Toronto, Ontario, Canada.,Institute of Medical Sciences University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Parks WT. Manifestations of Hypoxia in the Second and Third Trimester Placenta. Birth Defects Res 2017; 109:1345-1357. [DOI: 10.1002/bdr2.1143] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 09/21/2017] [Indexed: 02/01/2023]
Affiliation(s)
- W. Tony Parks
- Department of Pathology and Laboratory Medicine; Dartmouth-Hitchcock Medical Center; Lebanon New Hampshire
| |
Collapse
|
36
|
Ahn KH, Lee JH, Cho GJ, Hong SC, Oh MJ, Kim HJ. Placental thickness-to-estimated foetal weight ratios and small-for-gestational-age infants at delivery. J OBSTET GYNAECOL 2017. [PMID: 28631507 DOI: 10.1080/01443615.2017.1312306] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
This study aimed to determine the correlation between the placental thickness-to-estimated foetal weight ratio on midterm ultrasonography and small-for-gestational-age (SGA) infants. In this retrospective study, the placental thickness at the umbilical cord insertion site was measured and adjusted for foetal body weight at 18-24 weeks gestation. Investigators compared the data of women who delivered SGA infants (birth weight <10th percentile) with those of women who delivered non-SGA infants. Among the 1281 women in this study, those who delivered SGA infants were younger and less likely to be obese. Women with higher placental thickness-to-estimated foetal weight ratios delivered more SGA infants. In logistic regression analysis, a higher placental thickness-to-estimated foetal weight ratio remained associated with SGA infants. Since the placental thickness-to-estimated foetal weight ratio in midterm pregnancy was associated with infant body weight at delivery, this ratio could be an effective, adjunctive screening marker for predicting SGA status.
Collapse
Affiliation(s)
- Ki Hoon Ahn
- a Department of Obstetrics and Gynecology , Korea University College of Medicine , Seoul , Republic of Korea
| | - Joo Hak Lee
- a Department of Obstetrics and Gynecology , Korea University College of Medicine , Seoul , Republic of Korea
| | - Geum Joon Cho
- a Department of Obstetrics and Gynecology , Korea University College of Medicine , Seoul , Republic of Korea
| | - Soon-Cheol Hong
- a Department of Obstetrics and Gynecology , Korea University College of Medicine , Seoul , Republic of Korea
| | - Min-Jeong Oh
- a Department of Obstetrics and Gynecology , Korea University College of Medicine , Seoul , Republic of Korea
| | - Hai-Joong Kim
- a Department of Obstetrics and Gynecology , Korea University College of Medicine , Seoul , Republic of Korea
| |
Collapse
|
37
|
Robb KP, Cotechini T, Allaire C, Sperou A, Graham CH. Inflammation-induced fetal growth restriction in rats is associated with increased placental HIF-1α accumulation. PLoS One 2017; 12:e0175805. [PMID: 28423052 PMCID: PMC5397034 DOI: 10.1371/journal.pone.0175805] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 03/31/2017] [Indexed: 02/06/2023] Open
Abstract
Introduction Hypoxia-inducible factor 1-alpha (HIF-1α) is the oxygen-sensitive subunit of the transcription factor HIF-1, and its expression is increased in placentas from pregnancies complicated by pre-eclampsia (PE). Fetal growth restriction (FGR) and PE often share a common pathophysiology; however, it is unknown whether increased placental HIF-1α occurs in FGR. We previously demonstrated that aberrant maternal inflammation in rats resulted in altered utero-placental perfusion and FGR, both of which were prevented by administration of the nitric oxide mimetic glyceryl trinitrate (GTN). Our aim here was to determine whether abnormal maternal inflammation causing FGR is linked to placental HIF-1α accumulation and whether GTN administration could prevent increases in placental HIF-1α. Methods Levels of inflammatory factors in maternal plasma were measured using a multiplex assay after an injection of low-dose lipopolysaccharide (LPS) to rats on gestational day (GD) 13.5. Following three additional daily LPS injections from GD14.5–16.5, GD17.5 placentas were harvested for HIF-1α immunolocalisation; serial sections were also stained for the hypoxia marker pimonidazole. A subset of rats received LPS injections along with GTN delivered continuously (25 μg/h via a transdermal patch) on GD12.5-GD17.5. Results Within two hours of LPS administration, levels of maternal pro-inflammatory cytokines were increased compared with saline-treated controls. GD17.5 placentas of growth-restricted fetuses exhibited increased HIF-1α accumulation; however, this did not correlate with pimonidazole staining for which no differences were observed between groups. Furthermore, the LPS-mediated increases in maternal inflammatory cytokine levels and placental HIF-1α accumulation did not occur in rats treated with GTN. Discussion Our results demonstrate that inflammation-induced FGR is associated with increased placental HIF-1α accumulation; however, expression of this transcription factor may not correlate with regions of hypoxia in late-gestation placentas. The GTN-mediated attenuation of placental HIF-1α accumulation in LPS-treated rats provides insight into the mechanism by which GTN improves inflammation-induced complications of pregnancy.
Collapse
Affiliation(s)
- Kevin P. Robb
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Camille Allaire
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Arissa Sperou
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Charles H. Graham
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, Ontario, Canada
- * E-mail:
| |
Collapse
|
38
|
Yoshida A, Watanabe K, Iwasaki A, Kimura C, Matsushita H, Wakatsuki A. Placental oxidative stress and maternal endothelial function in pregnant women with normotensive fetal growth restriction. J Matern Fetal Neonatal Med 2017; 31:1051-1057. [PMID: 28367653 DOI: 10.1080/14767058.2017.1306510] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate the relationship between placental oxidative stress and maternal endothelial function in pregnant women with normotensive fetal growth restriction (FGR). METHODS We examined serum concentrations of oxygen free radicals (d-ROMs), maternal angiogenic factor (PlGF), and sFlt-1, placental oxidative DNA damage, and maternal endothelial function in 17 women with early-onset preeclampsia (PE), 18 with late-onset PE, 14 with normotensive FGR, and 21 controls. Flow-mediated vasodilation (FMD) was assessed as a marker of maternal endothelial function. Immunohistochemical analysis was performed to measure the proportion of placental trophoblast cell nuclei staining positive for 8-hydroxy-2'-deoxyguanosine (8-OHdG), a marker of oxidative DNA damage. RESULTS Maternal serum d-ROM, sFlt-1 concentrations, and FMD did not significantly differ between the control and normotensive FGR groups. The proportion of nuclei staining positive for 8-OHdG was significantly higher in the normotensive FGR group relative to the control group. CONCLUSIONS Our findings demonstrate that, despite the presence of placental oxidative DNA damage as observed in PE patients, pregnant women with normotensive FGR show no increase in the concentrations of sFlt-1 and d-ROMs, or a decrease in FMD.
Collapse
Affiliation(s)
- Atsumi Yoshida
- a Department of Obstetrics and Gynecology , Aichi Medical University School of Medicine , Nagakute , Aichi , Japan
| | - Kazushi Watanabe
- a Department of Obstetrics and Gynecology , Aichi Medical University School of Medicine , Nagakute , Aichi , Japan
| | - Ai Iwasaki
- a Department of Obstetrics and Gynecology , Aichi Medical University School of Medicine , Nagakute , Aichi , Japan
| | - Chiharu Kimura
- a Department of Obstetrics and Gynecology , Aichi Medical University School of Medicine , Nagakute , Aichi , Japan
| | - Hiroshi Matsushita
- a Department of Obstetrics and Gynecology , Aichi Medical University School of Medicine , Nagakute , Aichi , Japan
| | - Akihiko Wakatsuki
- a Department of Obstetrics and Gynecology , Aichi Medical University School of Medicine , Nagakute , Aichi , Japan
| |
Collapse
|
39
|
Wakeland AK, Soncin F, Moretto-Zita M, Chang CW, Horii M, Pizzo D, Nelson KK, Laurent LC, Parast MM. Hypoxia Directs Human Extravillous Trophoblast Differentiation in a Hypoxia-Inducible Factor-Dependent Manner. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:767-780. [PMID: 28167044 DOI: 10.1016/j.ajpath.2016.11.018] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 11/11/2016] [Accepted: 11/29/2016] [Indexed: 01/12/2023]
Abstract
Villous cytotrophoblasts are epithelial stem cells of the early human placenta, able to differentiate either into syncytiotrophoblasts in floating chorionic villi or extravillous trophoblasts (EVTs) at the anchoring villi. The signaling pathways regulating differentiation into these two lineages are incompletely understood. The bulk of placental growth and development in the first trimester occurs under low oxygen tension. One major mechanism by which oxygen regulates cellular function is through the hypoxia-inducible factor (HIF), a transcription factor complex stabilized under low oxygen tension to mediate cellular responses, including cell fate decisions. HIF is known to play a role in trophoblast differentiation in rodents; however, its role in human trophoblast differentiation is poorly understood. Using RNA profiling of sorted populations of primary first-trimester trophoblasts, we evaluated the first stage of EVT differentiation, the transition from epidermal growth factor receptor+ villous cytotrophoblasts into human leukocyte antigen-G+ proximal column EVT (pcEVT) and identified hypoxia as a major pcEVT-associated pathway. Using primary cytotrophoblasts, we determined that culture in low oxygen directs differentiation preferentially toward human leukocyte antigen-G+ pcEVT, and that an intact HIF complex is required for this process. Finally, using global RNA profiling, we identified integrin-linked kinase and associated cytoskeletal remodeling and adhesion to be among HIF-dependent pcEVT-associated signaling pathways. Taken together, we propose that oxygen regulates EVT differentiation through HIF-dependent modulation of various cell adhesion and morphology-related pathways.
Collapse
Affiliation(s)
- Anna K Wakeland
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Francesca Soncin
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Matteo Moretto-Zita
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Ching-Wen Chang
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Mariko Horii
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Don Pizzo
- Department of Pathology, University of California San Diego, La Jolla, California
| | - Katharine K Nelson
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California
| | - Louise C Laurent
- Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California; Department of Reproductive Medicine, University of California San Diego, La Jolla, California
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, California.
| |
Collapse
|
40
|
Karakus S, Bagci B, Bagci G, Sancakdar E, Yildiz C, Akkar O, Cetin A. SDF-1/CXCL12 and CXCR4 gene variants, and elevated serum SDF-1 levels are associated with preeclampsia. Hypertens Pregnancy 2016; 36:124-130. [PMID: 28001450 DOI: 10.1080/10641955.2016.1250904] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE We aimed to compare the frequencies of stromal cell-derived factor-1 (SDF-1) 3'A and CXCR4 single-nucleotide polymorphisms (SNPs) and serum SDF-1 levels in patients with preeclampsia (PE). METHODS In total, 89 women with PE and 89 control women were included in the study. Genotyping was done by polymerase chain reaction-restriction fragment length polymorphism method. Enzyme-linked immunosorbent assay method was used to measure serum SDF-1 level. RESULTS For SDF-1 3'A SNP, the frequency of GA genotype, total number of GA and AA genotypes, and the A allele frequency was higher in PE patients than controls (p = 0.04, 0.023, and 0.029, respectively). For CXCR4 SNP, the frequency of CT genotype, total number of CT and TT genotypes, and the T allele frequency were higher in PE patients than controls (p = 0.04, 0.006, and 0.005, respectively). SDF-1 serum level was detected higher in preeclamptic women compared with controls (p = 0.001). In PE patients, there was no significant association between serum SDF-1 levels and genotypes of SDF-1 3'A SNP. SDF-1 level was significantly higher in patients bearing CXCR4 CT genotype than CC genotype (p = 0.001). Furthermore, SDF-1 levels in patients bearing CT+TT genotype were found higher than that of patients with CC genotypes (p = 0.001). CONCLUSION Results of our study suggest that SDF-1 3'A and CXCR4 polymorphisms and elevated serum SDF-1 levels may have a role in the development of PE.
Collapse
Affiliation(s)
- Savas Karakus
- a Department of Obstetrics and Gynecology , Cumhuriyet University School of Medicine , Sivas , Turkey
| | - Binnur Bagci
- b Department of Nutrition and Dietetics , Cumhuriyet University School of Health Sciences , Sivas , Turkey
| | - Gokhan Bagci
- c Department of Medical Genetics , Cumhuriyet University School of Medicine , Sivas , Turkey
| | - Enver Sancakdar
- d Department of Biochemistry , Cumhuriyet University School of Medicine , Sivas , Turkey
| | - Caglar Yildiz
- a Department of Obstetrics and Gynecology , Cumhuriyet University School of Medicine , Sivas , Turkey
| | - Ozlem Akkar
- a Department of Obstetrics and Gynecology , Cumhuriyet University School of Medicine , Sivas , Turkey
| | - Ali Cetin
- a Department of Obstetrics and Gynecology , Cumhuriyet University School of Medicine , Sivas , Turkey
| |
Collapse
|
41
|
Sharma D, Sharma P, Shastri S. Genetic, metabolic and endocrine aspect of intrauterine growth restriction: an update. J Matern Fetal Neonatal Med 2016; 30:2263-2275. [DOI: 10.1080/14767058.2016.1245285] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Deepak Sharma
- Consultant Neonatologist, Department of Neonatology, NEOCLINIC, TN Mishra Marg, Everest Vihar, Nirman Nagar, Jaipur, Rajasthan, India,
| | - Pradeep Sharma
- Department of Medicine, Mahatma Gandhi Medical College, Jaipur, Rajasthan, India, and
| | - Sweta Shastri
- Department of Pathology, N.K.P Salve Medical College, Nagpur, Maharashtra, India
| |
Collapse
|
42
|
Abstract
OBJECTIVE To review the role of oxidative stress in two common placental-related disorders of pregnancy, miscarriage and preeclampsia. METHODS Review of published literature. RESULTS Miscarriage and preeclampsia manifest at contrasting stages of pregnancy, yet both have their roots in deficient trophoblast invasion during early gestation. Early after implantation, endovascular trophoblast cells migrate down the lumens of spiral arteries, and are associated with their physiological conversion into flaccid conduits. Initially these cells occlude the arteries, limiting maternal blood flow into the placenta. The embryo therefore develops in a low oxygen environment, protecting differentiating cells from damaging free radicals. Once embryogenesis is complete, the maternal intervillous circulation becomes fully established, and intraplacental oxygen concentration rises threefold. Onset of the circulation is normally a progressive periphery-center phenomenon, and high levels of oxidative stress in the periphery may induce formation of the chorion laeve. If trophoblast invasion is severely impaired, plugging of the spiral arteries is incomplete, and onset of the maternal intervillous circulation is premature and widespread throughout the placenta. Syncytiotrophoblastic oxidative damage is extensive, and likely a major contributory factor to miscarriage. Between these two extremes will be found differing degrees of trophoblast invasion compatible with ongoing pregnancy but resulting in deficient conversion of the spiral arteries and an ischemia-reperfusion-type phenomenon. Placental perfusion will be impaired to a greater or lesser extent, generating commensurate placental oxidative stress that is a major contributory factor to preeclampsia. CONCLUSION Miscarriage, missed miscarriage, and early- and late-onset preeclampsia represent a spectrum of disorders secondary to deficient trophoblast invasion.
Collapse
Affiliation(s)
- Graham J Burton
- Department of Anatomy, University of Cambridge, Cambridge, United Kingdom.
| | | |
Collapse
|
43
|
Endocannabinoids participate in placental apoptosis induced by hypoxia inducible factor-1. Apoptosis 2016; 21:1094-105. [DOI: 10.1007/s10495-016-1274-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
44
|
Zhao G, Miao H, Li X, Chen S, Hu Y, Wang Z, Hou Y. TGF-β3-induced miR-494 inhibits macrophage polarization via suppressing PGE2 secretion in mesenchymal stem cells. FEBS Lett 2016; 590:1602-13. [PMID: 27149081 DOI: 10.1002/1873-3468.12200] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/28/2016] [Accepted: 05/02/2016] [Indexed: 01/15/2023]
Abstract
Abnormal macrophage polarization at the maternal-fetal interface may contribute to the development of Preeclampsia (PE). The reason why macrophage polarization changed in PE is still unclear. Decidual mesenchymal stem cells (dMSCs) could regulate macrophage polarization. However, miRNA in dMSCs of PE were maladjusted. Therefore, we speculated that miRNA may affect dMSC-regulated macrophage polarization. In this study, we found that miR-494-overexpressed dMSCs inhibit M2 macrophage polarization and this inhibitory effect is mediated by miR-494-reduced PGE2 secretion. Furthermore, we proved that miR-494 is induced by TGF-β3. In summary, our findings suggest that the high expression of TGF-β3 in PE decidua stimulates miR-494 in dMSCs and attenuates the regulation of MSC switching the macrophage toward M2 type, contributing to an immune imbalance at maternal-fetal interface.
Collapse
Affiliation(s)
- Guangfeng Zhao
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China
| | - Huishuang Miao
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, China
| | - Xiujun Li
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China
| | - Shiwen Chen
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China
| | - Yali Hu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China
| | - Zhiqun Wang
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Nanjing University Medical School, China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School, Nanjing University, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing, China
| |
Collapse
|
45
|
Nüsken E, Wohlfarth M, Lippach G, Rauh M, Schneider H, Dötsch J, Nüsken KD. Reduced Perinatal Leptin Availability May Contribute to Adverse Metabolic Programming in a Rat Model of Uteroplacental Insufficiency. Endocrinology 2016; 157:1813-25. [PMID: 27007072 DOI: 10.1210/en.2015-1898] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Leptin availability in perinatal life critically affects metabolic programming. We tested the hypothesis that uteroplacental insufficiency and intrauterine stress affect perinatal leptin availability in rat offspring. Pregnant rats underwent bilateral uterine vessel ligation (LIG; n = 14), sham operation (SOP; n = 12), or no operation (controls, n = 14). Fetal livers (n = 180), placentas (n = 180), and maternal blood were obtained 4 hours (gestational day [E] 19), 24 hours (E20), and 72 hours (E22) after surgery. In the offspring, we took blood samples on E22 (n = 44), postnatal day (P) 1 (n = 29), P2 (n = 16), P7 (n = 30), and P12 (n = 30). Circulating leptin (ELISA) was significantly reduced in LIG (E22, P1, P2) and SOP offspring (E22). Postnatal leptin surge was delayed in LIG but was accelerated in SOP offspring. Placental leptin gene expression (quantitative RT-PCR) was reduced in LIG (E19, E20, E22) and SOP (E20, E22). Hepatic leptin receptor (Lepr-a, mediating leptin degradation) gene expression was increased in LIG fetuses (E20, E22) only. Surprisingly, hypoxia-inducible factors (Hif; Western blot) were unaltered in placentas and were reduced in the livers of LIG (Hif1a, E20; Hif2a, E19, E22) and SOP (Hif2a, E19) fetuses. Gene expression of prolyl hydroxylase 3, a factor expressed under hypoxic conditions contributing to Hif degradation, was increased in livers of LIG (E19, E20, E22) and SOP (E19) fetuses and in placentas of LIG and SOP (E19). In summary, reduced placental leptin production, increased fetal leptin degradation, and persistent perinatal hypoleptinemia are present in intrauterine growth restriction offspring, especially after uteroplacental insufficiency, and may contribute to perinatal programming of leptin resistance and adiposity in later life.
Collapse
Affiliation(s)
- Eva Nüsken
- Department of Pediatrics (E.N., M.W., G.L., J.D., K.-D.N.), Medical Faculty, University of Cologne, 50937 Cologne, Germany; and Department of Pediatrics (M.R., H.S.), University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Maria Wohlfarth
- Department of Pediatrics (E.N., M.W., G.L., J.D., K.-D.N.), Medical Faculty, University of Cologne, 50937 Cologne, Germany; and Department of Pediatrics (M.R., H.S.), University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Gregor Lippach
- Department of Pediatrics (E.N., M.W., G.L., J.D., K.-D.N.), Medical Faculty, University of Cologne, 50937 Cologne, Germany; and Department of Pediatrics (M.R., H.S.), University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics (E.N., M.W., G.L., J.D., K.-D.N.), Medical Faculty, University of Cologne, 50937 Cologne, Germany; and Department of Pediatrics (M.R., H.S.), University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Holm Schneider
- Department of Pediatrics (E.N., M.W., G.L., J.D., K.-D.N.), Medical Faculty, University of Cologne, 50937 Cologne, Germany; and Department of Pediatrics (M.R., H.S.), University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Jörg Dötsch
- Department of Pediatrics (E.N., M.W., G.L., J.D., K.-D.N.), Medical Faculty, University of Cologne, 50937 Cologne, Germany; and Department of Pediatrics (M.R., H.S.), University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Kai-Dietrich Nüsken
- Department of Pediatrics (E.N., M.W., G.L., J.D., K.-D.N.), Medical Faculty, University of Cologne, 50937 Cologne, Germany; and Department of Pediatrics (M.R., H.S.), University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| |
Collapse
|
46
|
Saunders ACE, McGonnigal B, Uzun A, Padbury J. The developmental expression of the CDK inhibitor p57(kip2) (Cdkn1c) in the early mouse placenta. Mol Reprod Dev 2016; 83:405-12. [PMID: 26988311 DOI: 10.1002/mrd.22637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 03/10/2016] [Indexed: 11/07/2022]
Abstract
p57(kip2) (encoded by the Cdkn1c gene) is a member of the cip/kip family of cyclin-dependent kinase inhibitors that mediates cell cycle arrest in G1, allowing cells to differentiate. In the placenta, p57(kip2) is involved in endoreduplication, formation of trophoblast giant cells, trophoblast invasion, and expansion of placental cell layers. Here, we quantitatively and qualitatively define the cell- and region-specific expression of mouse placental p57(kip2) using laser-capture microdissection, in situ hybridization, and immunohistochemistry. Cdkn1c RNA was quantified by real-time quantitative PCR. Co-expression of Pl1 was used to identify trophoblast giant cells while Tbpba was used to identify spongiotrophoblast cells. Timed sacrifices were also carried out at embryonic days E7.5, E8.5, E9.5, and E12.5 to profile the expression in embryos and their placentas. At E8.5, intense expression of Cdkn1c was seen in invasive TGCs and the ectoplacental cone. Cdkn1c expression was more diffuse and more abundant in the labyrinth that in the junctional zone at both E9.5 and E12.5. Immunohistochemistry revealed robust p57(kip2) staining in trophoblast giant cells and in the ectoplacental cone at E8.5. p57(kip2) protein was seen in giant cells and throughout the labyrinth, although its abundance was reduced in the junctional zone at E9.5, and became more diffuse by E12.5. The early and intense expression in trophoblast giant cells is consistent with a role for p57(kip2) in the invasive phenotype of these cells. Mol. Reprod. Dev. 83: 405-412, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ann Catherine Eugenia Saunders
- Department of Pediatrics, Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island.,Division of Biology and Medicine Graduate Program in Pathobiology, Brown University Providence, Providence, Rhode Island
| | - Bethany McGonnigal
- Department of Pediatrics, Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island
| | - Alper Uzun
- Department of Pediatrics, Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island
| | - James Padbury
- Department of Pediatrics, Brown University, Women and Infants Hospital of Rhode Island, Providence, Rhode Island.,Division of Biology and Medicine Graduate Program in Pathobiology, Brown University Providence, Providence, Rhode Island
| |
Collapse
|
47
|
Dynamic regulation of HIF1Α stability by SUMO2/3 and SENP3 in the human placenta. Placenta 2016; 40:8-17. [PMID: 27016777 DOI: 10.1016/j.placenta.2016.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/26/2016] [Accepted: 02/03/2016] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Hypoxia-inducible factor 1A (HIF1A) stability is tightly regulated by hydroxylation and ubiquitination. Emerging evidence indicates that HIF1A is also controlled by the interplay between SUMO-specific ligases, which execute protein SUMOylation, and Sentrin/SUMO-specific proteases that de-SUMOylate target proteins. Given the significance of HIF1A in the human placenta, we investigated whether placental HIF1A is subject to SUMOylation in physiological and pathological conditions. METHODS Placentae were obtained from normal and pregnancies complicated by preeclampsia. Human choriocarcinoma JEG3 cells were maintained at either 21% or 3% oxygen or exposed to sodium nitroprusside (SNP). Cells were transfected with SUMO2/3 constructs with and without Mg132, a proteasome inhibitor. Expression, distribution and associations of SUMO/SENPs and HIF1A were evaluated by Western blotting, immunohistochemistry and co-immunoprecipitation. RESULTS HIF1A-SUMO2/3 associations peaked at 9-10 weeks, while its deSUMOylation by SENP3 was greatest at 10-12 weeks. In E-PE, HIF1A deSUMOylation by SENP3 was significantly elevated, while HIF1A-SUMO2/3 associations remained constant. In vitro, overexpression of SUMO2/3 de-stabilized HIF1A in hypoxia, and abrogated HIF1A expression following Mg132 treatment in normoxia. Hypoxia and SNP treatments promoted SENP3 nuclear redistribution from nucleoli to the nucleoplasm where it associates with HIF1A. CONCLUSION During early placental development, SUMOylation events control HIF1A stability in an oxygen-dependent manner. In E-PE, enhanced deSUMOylation of HIF1A by SENP3 may in part contribute to increased HIF1A activity and stability found in this pathology.
Collapse
|
48
|
Kimball R, Wayment M, Merrill D, Wahlquist T, Reynolds PR, Arroyo JA. Hypoxia reduces placental mTOR activation in a hypoxia-induced model of intrauterine growth restriction (IUGR). Physiol Rep 2015; 3:3/12/e12651. [PMID: 26660559 PMCID: PMC4760431 DOI: 10.14814/phy2.12651] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) is a protein that regulates cell growth in response to altered nutrient and growth factor availability. Our objective was to assess activated mTOR and its intracellular intermediates p70, and 4EBP1 in placental and invasive trophoblast cells in a hypoxia‐induced model of intrauterine growth restriction (IUGR) in rats. Rats were treated with hypoxia (9%) for 4 days. Placental and fetal weights, as well as conceptus numbers were recorded at the time of necropsy. Immunohistochemistry was used to determine the level of trophoblast invasion and apoptosis. Western blots were used to determine the activation of mTOR, p70, and 4EBP1 in the placenta and the uterine mesometrial compartment. We observed (1) decreased placental (21%) and fetal (24%) weights (P < 0.05); (2) decreased trophoblast invasion; (3) significantly increased active 4EBP1 (28%; P < 0.05) in invasive trophoblast cells yet no changes in the activation of mTOR and p70 proteins; and (4) a significant decrease in the activation of mTOR (48%; P < 0.05) with no differences in p70 or 4EBP1 activation in the placenta. We conclude that the development of IUGR is correlated with decreased activation of the mTOR protein in the placenta and increased 4EBP1 activity in the invading trophoblast. These results provide important insight into the physiological relevance of these pathways. Furthermore, modification of these and other related targets during gestation may alleviate IUGR severity.
Collapse
Affiliation(s)
- Rebecca Kimball
- Lung and Placenta Research Laboratory, , Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Montana Wayment
- Lung and Placenta Research Laboratory, , Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Daniel Merrill
- Lung and Placenta Research Laboratory, , Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Tyler Wahlquist
- Lung and Placenta Research Laboratory, , Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Paul R Reynolds
- Lung and Placenta Research Laboratory, , Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| | - Juan A Arroyo
- Lung and Placenta Research Laboratory, , Physiology and Developmental Biology, Brigham Young University, Provo, Utah
| |
Collapse
|
49
|
Perez-Sepulveda A, Monteiro LJ, Dobierzewska A, España-Perrot PP, Venegas-Araneda P, Guzmán-Rojas AM, González MI, Palominos-Rivera M, Irarrazabal CE, Figueroa-Diesel H, Varas-Godoy M, Illanes SE. Placental Aromatase Is Deficient in Placental Ischemia and Preeclampsia. PLoS One 2015; 10:e0139682. [PMID: 26444006 PMCID: PMC4596497 DOI: 10.1371/journal.pone.0139682] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/16/2015] [Indexed: 12/03/2022] Open
Abstract
Introduction Preeclampsia is a maternal hypertensive disorder with uncertain etiology and a leading cause of maternal and fetal mortality worldwide, causing nearly 40% of premature births delivered before 35 weeks of gestation. The first stage of preeclampsia is characterized by reduction of utero-placental blood flow which is reflected in high blood pressure and proteinuria during the second half of pregnancy. In human placenta androgens derived from the maternal and fetal adrenal glands are converted into estrogens by the enzymatic action of placental aromatase. This implies that alterations in placental steroidogenesis and, subsequently, in the functionality or bioavailability of placental aromatase may be mechanistically involved in the pathophysiology of PE. Methods Serum samples were collected at 32–36 weeks of gestation and placenta biopsies were collected at time of delivery from PE patients (n = 16) and pregnant controls (n = 32). The effect of oxygen tension on placental cells was assessed by incubation JEG–3 cells under 1% and 8% O2 for different time periods, Timed-mated, pregnant New Zealand white rabbits (n = 6) were used to establish an in vivo model of placental ischemia (achieved by ligature of uteroplacental vessels). Aromatase content and estrogens and androgens concentrations were measured. Results The protein and mRNA content of placental aromatase significantly diminished in placentae obtained from preeclamptic patients compared to controls. Similarly, the circulating concentrations of 17-β-estradiol/testosterone and estrone/androstenedione were reduced in preeclamptic patients vs. controls. These data are consistent with a concomitant decrease in aromatase activity. Aromatase content was reduced in response to low oxygen tension in the choriocarcinoma JEG–3 cell line and in rabbit placentae in response to partial ligation of uterine spiral arteries, suggesting that reduced placental aromatase activity in preeclamptic patients may be associated with chronic placental ischemia and hypoxia later in gestation. Conclusions Placental aromatase expression and functionality are diminished in pregnancies complicated by preeclampsia in comparison with healthy pregnant controls.
Collapse
Affiliation(s)
- Alejandra Perez-Sepulveda
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Lara J. Monteiro
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Aneta Dobierzewska
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Pedro P. España-Perrot
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Pía Venegas-Araneda
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Alejandra M. Guzmán-Rojas
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - María I. González
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Macarena Palominos-Rivera
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Carlos E. Irarrazabal
- Laboratory of Molecular Physiology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Horacio Figueroa-Diesel
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
- Perinatal Unit, Clínica Dávila, Santiago, Chile
| | - Manuel Varas-Godoy
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
| | - Sebastián E. Illanes
- Department of Obstetrics and Gynecology, Laboratory of Reproductive Biology, Faculty of Medicine, Universidad de Los Andes, Santiago, Chile
- Perinatal Unit, Clínica Dávila, Santiago, Chile
- * E-mail:
| |
Collapse
|
50
|
Ornaghi S, Mueller M, Barnea ER, Paidas MJ. Thrombosis during pregnancy: Risks, prevention, and treatment for mother and fetus-harvesting the power of omic technology, biomarkers and in vitro or in vivo models to facilitate the treatment of thrombosis. ACTA ACUST UNITED AC 2015; 105:209-25. [DOI: 10.1002/bdrc.21103] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sara Ornaghi
- Department of Obstetrics and Gynecology; University of Milan-Bicocca; Monza Italy
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale Women and Children's Center for Blood Disorders and Preeclampsia Advancement, Yale University School of Medicine; New Haven Connecticut
| | - Martin Mueller
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale Women and Children's Center for Blood Disorders and Preeclampsia Advancement, Yale University School of Medicine; New Haven Connecticut
- Department of Obstetrics and Gynecology; University Hospital Bern; Bern Switzerland
| | - Eytan R. Barnea
- Society for the Investigation of Early Pregnancy; Cherry Hill New Jersey
- BioIncept LLC; Cherry Hill New Jersey
| | - Michael J. Paidas
- Department of Obstetrics, Gynecology and Reproductive Sciences; Yale Women and Children's Center for Blood Disorders and Preeclampsia Advancement, Yale University School of Medicine; New Haven Connecticut
| |
Collapse
|