1
|
Grah M, Poljak L, Starčević M, Stanojević M, Vukojević K, Saraga-Babić M, Salihagić AK. Does placental VEGF-A protein expression predict early neurological outcome of neonates from FGR complicated pregnancies? J Perinat Med 2024; 52:783-792. [PMID: 39028860 DOI: 10.1515/jpm-2024-0138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/03/2024] [Indexed: 07/21/2024]
Abstract
OBJECTIVES Fetal hypoxia due to placental dysfunction is the hallmark of fetal growth restriction (FGR). Preferential perfusion of the brain (brain-sparing effect), as a part of physiological placental cardiovascular compensatory mechanisms to hypoxia, in FGR was reported. Therefore, the correlation between vascular endothelial growth factor A (VEGF-A) protein expression in the FGR placentas and newborns' early neurological outcome was examined. METHODS This study included 50 women with FGR complicated pregnancies and 30 uneventful pregnancies. Fetal hemodynamic parameters, neonatal acid-base status after delivery, placental pathohistology and VEGF-A expression were followed. Early neonatal morphological brain evaluation by ultrasound and functional evaluation of neurological status by Amiel - Tison Neurological Assessment at Term (ATNAT) were performed. RESULTS VEGF-A protein expression level was significantly higher in the FGR placentas than normal term placentas (Fisher-Freeman-Halton's test, p≤0.001). No statistically significant correlation between placental VEGF-A expression and different prenatal and postnatal parameters was noticed. Whereas the alteration of an early neurological status assessed by ATNAT was found in 58 % of FGR newborns, morphological brain changes evaluated by UZV was noticed in 48 % of cases. No association between the level of placental VEGF-A expression and the early neurological deficits was found. CONCLUSIONS As far as we know this is the first study of a possible connection between VEGF-A protein expression in the FGR placentas and neonates' early neurological outcomes. The lack of correlation between the FGR placental VEGF-A expression and neonates' neurological outcome could indicate that optimal early neurodevelopment may take place due to compensatory mechanism not related to placental VEGF-A expression.
Collapse
Affiliation(s)
- Maja Grah
- Department of Obstetrics and Gynecology, Clinical Hospital "Sveti Duh", School of Medicine, 37632 University of Zagreb , Zagreb, Croatia
| | - Ljiljana Poljak
- Department of Physiology, School of Medicine, 37632 University of Zagreb , Zagreb, Croatia
| | - Mirta Starčević
- Division of Neonatology, Department of Gynecology and Obstetrics, Clinical Hospital Center "Zagreb", Zagreb, Croatia
| | - Milan Stanojević
- Neonatal Unit, Department of Obstetrics and Gynecology, Clinical Hospital "Sveti Duh", School of Medicine, 37632 University of Zagreb , Zagreb, Croatia
| | - Katarina Vukojević
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Split, Croatia
| | - Mirna Saraga-Babić
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Split, Croatia
| | - Aida Kadić Salihagić
- Department of Physiology, School of Medicine, 37632 University of Zagreb , Zagreb, Croatia
| |
Collapse
|
2
|
Mrozikiewicz AE, Kurzawińska G, Walczak M, Skrzypczak-Zielińska M, Ożarowski M, Jędrzejczak P. Up-regulated mRNA expression of VEGFA receptors (FLT1 and KDR) in placentas after assisted reproductive technology fertilization. J Appl Genet 2024; 65:531-540. [PMID: 38238614 PMCID: PMC11310245 DOI: 10.1007/s13353-023-00823-2] [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: 10/25/2023] [Revised: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 08/09/2024]
Abstract
Placental angiogenesis is a pivotal process for feto-maternal circulation and ensures efficient development of the placenta throughout pregnancy. Many factors during in vitro fertilization and embryo transfer procedures may affect placental gene expression and fetus development. The present study aimed to identify differences in angiogenesis-related gene (VEGFA, FGF2, FLT1, and KDR) expression profiles in placentas after assisted reproductive technology fertilization and natural conception in healthy women. In a case-control study, term placentas were collected from Caucasian women after assisted reproductive technology fertilization (N = 20) and after natural conception in women with uncomplicated pregnancy (N = 9). The mRNA expression in placentas was examined for VEGFA, FGF2, FLT1, and KDR genes by real-time quantitative polymerase chain reaction (RT-qPCR). Group stratification was performed for comparison of investigated genes between the type of embryo transferred (fresh/frozen), place of tissue donation (center/margin), and newborns' gender (male/female). In the ART placentas, significant down-regulation of VEGFA gene (p = 0.016) and up-regulation of FLT1 (p = 0.026) and KDR (p < 0.001) gene receptors were observed. Genes encoding VEGFA receptors were up-regulated in both fresh (ET) and frozen (FET) embryo transfer groups compared to controls. For the FLT1 gene, a statistically significant difference was observed between the frozen embryo transfer group and the controls (p = 0.032). Relative expression of KDR was significantly higher for both embryo transfer groups compared to controls (p < 0.001) and between ET and FET (p = 0.002). No statistically significant differences were observed between placental expression in different places of tissue donation and newborns' gender. We observed differences in the placental expression of VEGFA and its receptors FLT1 and KDR in pregnancies after assisted reproductive technology compared to naturally conceived pregnancies. More research is needed to clarify these alterations that may affect placental development and fetal health.
Collapse
Affiliation(s)
- Aleksandra E Mrozikiewicz
- Department of Obstetrics and Women's Diseases, Poznan University of Medical Sciences, Polna 33, 60-535, Poznan, Poland
| | - Grażyna Kurzawińska
- Division of Perinatology and Women's Diseases, Poznan University of Medical Sciences, Polna 33, 60-535, Poznan, Poland
- Laboratory of Molecular Biology in Division of Perinatology and Women's Diseases, Poznan University of Medical Sciences, Polna 33, 60-535, Poznan, Poland
| | - Michał Walczak
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszyńska 32, 60-479, Poznan, Poland
| | | | - Marcin Ożarowski
- Department of Biotechnology, Institute of Natural Fibres and Medicinal Plants, National Research Institute, 60-630, Poznan, Poland.
| | - Piotr Jędrzejczak
- Department of Infertility and Reproductive Endocrinology, Poznan University of Medical Sciences, Polna 33, 60-535, Poznan, Poland
| |
Collapse
|
3
|
Umapathy A, Clark A, Sehgal A, Karanam V, Rajaraman G, Kalionis B, Jones H, James J, Murthi P. Molecular regulators of defective placental and cardiovascular development in fetal growth restriction. Clin Sci (Lond) 2024; 138:761-775. [PMID: 38904187 PMCID: PMC11193155 DOI: 10.1042/cs20220428] [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: 12/29/2023] [Revised: 05/12/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
Placental insufficiency is one of the major causes of fetal growth restriction (FGR), a significant pregnancy disorder in which the fetus fails to achieve its full growth potential in utero. As well as the acute consequences of being born too small, affected offspring are at increased risk of cardiovascular disease, diabetes and other chronic diseases in later life. The placenta and heart develop concurrently, therefore placental maldevelopment and function in FGR may have profound effect on the growth and differentiation of many organ systems, including the heart. Hence, understanding the key molecular players that are synergistically linked in the development of the placenta and heart is critical. This review highlights the key growth factors, angiogenic molecules and transcription factors that are common causes of defective placental and cardiovascular development.
Collapse
Affiliation(s)
- Anandita Umapathy
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Alys Clark
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
- Auckland Bioengineering Institute, Bioengineering Institute, New Zealand
| | - Arvind Sehgal
- Department of Paediatrics, Monash University, Melbourne, VIC, Australia and Monash Newborn, Monash Children’s Hospital, Melbourne, VIC, Australia
| | - Vijaya Karanam
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
| | - Gayathri Rajaraman
- First year college, Victoria University, St Albans, Victoria 3021, Australia
| | - Bill Kalionis
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Victoria, Australia
| | - Helen N. Jones
- Department of Physiology and Aging, University of Florida College of Medicine, Gainesville, FL, U.S.A
- Center for Research in Perinatal Outcomes, University of Florida College of Medicine, Gainesville, FL, U.S.A
| | - Jo James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
- Auckland Bioengineering Institute, Bioengineering Institute, New Zealand
| | - Padma Murthi
- Department of Obstetrics, Gynaecology and Newborn Health, University of Melbourne and Royal Women’s Hospital, Victoria, Australia
- Department of Maternal Fetal Medicine, Pregnancy Research Centre, Royal Women’s Hospital, Victoria, Australia
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
4
|
Frolova ER, Tumanova UN, Gladkova KA, Khodzhaeva ZS, Shchegolev AI. The Number of Syncytial Knots and the Intensity of VEGF Expression in the Villi of the Monochorionic Diamniotic Placenta in Selective Fetal Growth Restriction. Bull Exp Biol Med 2024; 177:389-394. [PMID: 39134814 DOI: 10.1007/s10517-024-06195-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Indexed: 08/28/2024]
Abstract
A comparative study was carried out to analyze the number of syncytial knots and the intensity of expression of vascular endothelial growth factor (VEGF) in the villi of the monochorionic diamniotic placenta in pregnancies complicated by the syndrome of selective fetal growth restriction (sFGR). We performed a morphological analysis of 32 monochorionic diamniotic placentas after term delivery which were divided into two groups. The main group included the placentas of 15 puerperas whose pregnancies were complicated by sFGR. The control group included twin placentas of 17 puerperas without signs of sFGR. The number of syncytial knots was determined by histological studies, and the levels of VEGF expression in syncytiotrophoblast and capillary endotheliocytes of the placental villi were determined by immunohistochemical studies. The study showed an increase in the number of syncytial knots in the villi of the placental part of the fetus with sFGR which indicated the development of preplacental hypoxia. A significant increase in the level of VEGF expression in the syncytiotrophoblast and vascular endothelium of the villi should be considered as a manifestation of a compensatory adaptational response to hypoxia, though it is insufficient to prevent the development of sFGR.
Collapse
Affiliation(s)
- E R Frolova
- National Medical Research Center of Obstetrics, Gynecology, and Perinatology named after Academician V.I.Kulakov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - U N Tumanova
- National Medical Research Center of Obstetrics, Gynecology, and Perinatology named after Academician V.I.Kulakov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - K A Gladkova
- National Medical Research Center of Obstetrics, Gynecology, and Perinatology named after Academician V.I.Kulakov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Z S Khodzhaeva
- National Medical Research Center of Obstetrics, Gynecology, and Perinatology named after Academician V.I.Kulakov, Ministry of Health of the Russian Federation, Moscow, Russia
| | - A I Shchegolev
- National Medical Research Center of Obstetrics, Gynecology, and Perinatology named after Academician V.I.Kulakov, Ministry of Health of the Russian Federation, Moscow, Russia.
| |
Collapse
|
5
|
Tamil Barathi P, Mohanapriya A. Pre-eclampsia: Re-visiting pathophysiology, role of immune cells, biomarker identification and recent advances in its management. J Reprod Immunol 2024; 163:104236. [PMID: 38555746 DOI: 10.1016/j.jri.2024.104236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024]
Abstract
Pre-eclampsia (PE) is a hypertension condition that occurs exclusively during pregnancy and has the potential to impact nearly all organ systems. It is estimated to complicate approximately 2-8% of pregnancies worldwide. PE is a prominent medical disorder that poses a significant risk to pregnant mothers and their infants. This review commences by giving the most up-to- date concepts about the pathophysiology of PE. The condition involves atypical infiltration of trophoblast cells into the spiral arteries of the decidua and myometrium, resulting in an insufficient establishment of proper blood flow between the uterus and placenta. The aberrant activation of natural killer (NK) cells in both the peripheral blood and the decidua has been identified as one of the contributing factors to the development of PE. The strong evidence for the genetic etiology of PE is provided by the association between maternal killer cell immunoglobulin-like receptor (KIR) and Human Leukocyte Antigen (HLA-C) in trophoblast cells. Recent observations provide evidence that changes in the expression of anti-angiogenic factors in the placenta are the underlying cause of the clinical symptoms associated with the condition. This review also provides a comprehensive overview of the latest advancements in understanding the underlying causes of PE. It specifically highlights the emergence of new diagnostic biomarkers and their potential implications for therapeutic interventions in managing this medical condition.
Collapse
Affiliation(s)
- Palanisamy Tamil Barathi
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| | - Arumugam Mohanapriya
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India.
| |
Collapse
|
6
|
AbdelFattah S, Morsy M, Ahmed AM, Abdelsalam H, Hosny G. Microcellular approach for the pathogenesis of placenta accreta spectrum inflammatory versus apoptotic pathways; a thorough look on Treg, dNK and VEGF. Pathol Res Pract 2024; 254:155153. [PMID: 38301363 DOI: 10.1016/j.prp.2024.155153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 02/03/2024]
Abstract
Placenta accreta spectrum (PAS) is a disorder of irregular placental invasion to the surrounding structures, it is a leading cause of maternal morbidity and mortality. This study was theorized to perceive the role of Treg cells and VEGF which appealed to play a role in the pathogenesis of nonstandard extreme placental invasion. The study was carried out on 40 pregnant women; Group I (control group), and Group II (placenta accrete spectrum PAS). Light microscopic, immune-histochemical; CD 56 (NK CELLS) and CD 45 RO (T reg) western blot; P53 and VEGF morphometry and statistical analysis were done. H&E-stained sections revealed Placental tissue in unswerving contact with the myometrium, deficient decidual layer, hemorrhage, congested edematous blood vessels. The mean area percent of collagen, Treg, P53, and VEGF exposed a significant increase in the placenta accreta group when compared to that of control women. Nonetheless, the mean area percent of NK cells displayed a significant decrement PAS cases are associated with low levels of NK cells and increased levels of Treg cells, P 53, and VEGF, promoting the hyperinvasive behavior of trophoblasts of placenta accreta and dysregulate placental vascular remodeling.
Collapse
Affiliation(s)
- Shereen AbdelFattah
- Anatomy and Embryology, Kasralainy, Faculty of Medicine, Cairo University, Cairo, Egypt.
| | - Medhat Morsy
- Anatomy and Embryology, Kasralainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed Mahmoud Ahmed
- Obstetrics and Gynecology, Kasralainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Haitham Abdelsalam
- Anatomy and Embryology, Kasralainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Gamal Hosny
- Anatomy and Embryology, Kasralainy, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
7
|
Yin T, Li X, Li Y, Zang X, Liu L, Du M. Macrophage plasticity and function in cancer and pregnancy. Front Immunol 2024; 14:1333549. [PMID: 38274812 PMCID: PMC10808357 DOI: 10.3389/fimmu.2023.1333549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
As the soil of life, the composition and shaping process of the immune microenvironment of the uterus is worth exploring. Macrophages, indispensable constituents of the innate immune system, are essential mediators of inflammation and tissue remodeling as well. Recent insights into the heterogeneity of macrophage subpopulations have renewed interest in their functional diversity in both physiological and pathological settings. Macrophages display remarkable plasticity and switch from one phenotype to another. Intrinsic plasticity enables tissue macrophages to perform a variety of functions in response to changing tissue contexts, such as cancer and pregnancy. The remarkable diversity and plasticity make macrophages particularly intriguing cells given their dichotomous role in either attacking or protecting tumors and semi-allogeneic fetuses, which of both are characterized functionally by immunomodulation and neovascularization. Here, we reviewed and compared novel perspectives on macrophage biology of these two settings, including origin, phenotype, differentiation, and essential roles in corresponding microenvironments, as informed by recent studies on the heterogeneity of macrophage identity and function, as well as their mechanisms that might offer opportunities for new therapeutic strategies on malignancy and pregnancy complications.
Collapse
Affiliation(s)
- Tingxuan Yin
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xinyi Li
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yanhong Li
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Lu Liu
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Meirong Du
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
8
|
S C, G G, LA S, W N, P M, L A, A W, V F, P W, D G, T BT. Transcriptomic profiling reveals differential cellular response to copper oxide nanoparticles and polystyrene nanoplastics in perfused human placenta. ENVIRONMENT INTERNATIONAL 2023; 177:108015. [PMID: 37315489 DOI: 10.1016/j.envint.2023.108015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/16/2023]
Abstract
The growing nanoparticulate pollution (e.g. engineered nanoparticles (NPs) or nanoplastics) has been shown to pose potential threats to human health. In particular, sensitive populations such as pregnant women and their unborn children need to be protected from harmful environmental exposures. However, developmental toxicity from prenatal exposure to pollution particles is not yet well studied despite evidence of particle accumulation in human placenta. Our study aimed to investigate how copper oxide NPs (CuO NPs; 10-20 nm) and polystyrene nanoplastics (PS NPs; 70 nm) impact on gene expression in ex vivo perfused human placental tissue. Whole genome microarray analysis revealed changes in global gene expression profile after 6 h of perfusion with sub-cytotoxic concentrations of CuO (10 µg/mL) and PS NPs (25 µg/mL). Pathway and gene ontology enrichment analysis of the differentially expressed genes suggested that CuO and PS NPs trigger distinct cellular response in placental tissue. While CuO NPs induced pathways related to angiogenesis, protein misfolding and heat shock responses, PS NPs affected the expression of genes related to inflammation and iron homeostasis. The observed effects on protein misfolding, cytokine signaling, and hormones were corroborated by western blot (accumulation of polyubiquitinated proteins) or qPCR analysis. Overall, the results of the present study revealed extensive and material-specific interference of CuO and PS NPs with placental gene expression from a single short-term exposure which deserves increasing attention. In addition, the placenta, which is often neglected in developmental toxicity studies, should be a key focus in the future safety assessment of NPs in pregnancy.
Collapse
Affiliation(s)
- Chortarea S
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Gupta G
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Saarimäki LA
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Netkueakul W
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Manser P
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Aengenheister L
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland; Human Biomonitoring Research Unit, Department of Precision Health, Luxembourg Institute of Health (LIH), 1 A-B, Rue Thomas Edison, L-1445 Strassen, Luxembourg
| | - Wichser A
- Laboratory for Advanced Analytical Technologies, Empa, Swiss Federal Laboratories for Materials, Science and Technology, Dübendorf, Switzerland
| | - Fortino V
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, Kuopio, Finland
| | - Wick P
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland
| | - Greco D
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Buerki-Thurnherr T
- Laboratory for Particles-Biology Interactions, Swiss Federal Laboratories for Materials Science and Technology (Empa), 9014 St. Gallen, Switzerland.
| |
Collapse
|
9
|
Zhang Y, Liu Z, Sun H. Fetal-maternal interactions during pregnancy: a 'three-in-one' perspective. Front Immunol 2023; 14:1198430. [PMID: 37350956 PMCID: PMC10282753 DOI: 10.3389/fimmu.2023.1198430] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 05/24/2023] [Indexed: 06/24/2023] Open
Abstract
A successful human pregnancy requires the maternal immune system to recognize and tolerate the semi-allogeneic fetus, allowing for appropriate trophoblasts invasion and protecting the fetus from invading pathogens. Therefore, maternal immunity is critical for the establishment and maintenance of pregnancy, especially at the maternal-fetal interface. Anatomically, the maternal-fetal interface has both maternally- and fetally- derived cells, including fetal originated trophoblasts and maternal derived immune cells and stromal cells. Besides, a commensal microbiota in the uterus was supposed to aid the unique immunity in pregnancy. The appropriate crosstalk between fetal derived and maternal originated cells and uterine microbiota are critical for normal pregnancy. Dysfunctional maternal-fetal interactions might be associated with the development of pregnancy complications. This review elaborates the latest knowledge on the interactions between trophoblasts and decidual immune cells, highlighting their critical roles in maternal-fetal tolerance and pregnancy development. We also characterize the role of commensal bacteria in promoting pregnancy progression. Furthermore, this review may provide new thought on future basic research and the development of clinical applications for pregnancy complications.
Collapse
Affiliation(s)
- Yonghong Zhang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhaozhao Liu
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
10
|
Macías-Salas A, Sosa-Macías M, Barragán-Zúñiga LJ, Blanco-Castañeda R, Damiano A, Garcia-Robles R, Ayala-Ramírez P, Bueno-Sánchez J, Giachini FR, Escudero C, Galaviz-Hernández C. Preeclampsia association of placental nucleotide variations in eNOS, VEGFA, and FLT-1 genes in Latin American pregnant women. Placenta 2023; 135:1-6. [PMID: 36878143 DOI: 10.1016/j.placenta.2023.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/06/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023]
Abstract
INTRODUCTION Preeclampsia is a leading cause of maternal and fetal morbidity in low- and middle-income countries, including those in Latin America. Placental vascular alterations are crucial in the pathophysiology of preeclampsia and few studies have evaluated nucleotide variations on genes associated with vascular regulation in the human placenta. This study aimed to evaluate whether placental nucleotide variations on eNOS, VEGFA, and FLT-1 genes are more frequently associated with preeclampsia in the Latin American population. METHODS This case-control study included placental tissue from 88 controls and 82 cases that were genotyped through Taqman probes for eNOS, VEGFA, and FLT-1 genes. The intergroup comparisons were analyzed with the Mann-Whitney U test. Genotype and allele frequencies were compared by the X2 test. The association between the nucleotide variants with preeclampsia was evaluated through logistic regression analysis. RESULTS A significant association was observed for VEGFA SNV rs2010963 (OR 1.95; CI 95% 1.13-3.37), after adjusting for population substructure. The allele combination T, G, G, C, C, C (rs2070744, rs1799983, rs2010963, rs3025039, rs699947 and rs4769613 respectively), showed a negative association with preeclampsia (OR 0.08; CI 95% 0.01-0.93). RESULTS DISCUSSION Placental SNV rs2010963 in the VEGFA gene was a risk factor for preeclampsia, while the allele combination T, G, G, C, C, C may represent potential protective factors for preeclampsia within Latin American women.
Collapse
Affiliation(s)
- Alejo Macías-Salas
- Pharmacogenomics Academia, Instituto Politécnico Nacional-CIIDIR Durango, Mexico
| | - Martha Sosa-Macías
- Pharmacogenomics Academia, Instituto Politécnico Nacional-CIIDIR Durango, Mexico; Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chillan, Chile
| | | | | | - Alicia Damiano
- Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chillan, Chile; Laboratorio de Biología de la Reproducción, IFIBIO Houssay-UBA- CONICET, Buenos Aires, Argentina; Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Reggie Garcia-Robles
- Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chillan, Chile; Department of Physiological Sciences, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Paola Ayala-Ramírez
- Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chillan, Chile; Human Genetics Institute, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Julio Bueno-Sánchez
- Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chillan, Chile; Grupo Reproducción, Department of Obstetrics and Gynecology, Department of Physiology and biochemisty, Universidad de Antioquia, Medellín, Colombia
| | - Fernanda Regina Giachini
- Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chillan, Chile; Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Carlos Escudero
- Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chillan, Chile; Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile.
| | - Carlos Galaviz-Hernández
- Pharmacogenomics Academia, Instituto Politécnico Nacional-CIIDIR Durango, Mexico; Red Iberoamericana de Alteraciones Vasculares en Transtornos del Embarazo (RIVATREM), Chillan, Chile.
| |
Collapse
|
11
|
Zhu Y, Liu X, Xu Y, Lin Y. Hyperglycemia disturbs trophoblast functions and subsequently leads to failure of uterine spiral artery remodeling. Front Endocrinol (Lausanne) 2023; 14:1060253. [PMID: 37091848 PMCID: PMC10113679 DOI: 10.3389/fendo.2023.1060253] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
Uterine spiral artery remodeling is necessary for fetal growth and development as well as pregnancy outcomes. During remodeling, trophoblasts invade the arteries, replace the endothelium and disrupt the vascular smooth muscle, and are strictly regulated by the local microenvironment. Elevated glucose levels at the fetal-maternal interface are associated with disorganized placental villi and poor placental blood flow. Hyperglycemia disturbs trophoblast proliferation and invasion via inhibiting the epithelial-mesenchymal transition, altering the protein expression of related proteases (MMP9, MMP2, and uPA) and angiogenic factors (VEGF, PIGF). Besides, hyperglycemia influences the cellular crosstalk between immune cells, trophoblast, and vascular cells, leading to the failure of spiral artery remodeling. This review provides insight into molecular mechanisms and signaling pathways of hyperglycemia that influence trophoblast functions and uterine spiral artery remodeling.
Collapse
Affiliation(s)
- Yueyue Zhu
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Xiaorui Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yichi Xu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Yi Lin
- Reproductive Medicine Center, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yi Lin,
| |
Collapse
|
12
|
Placental Morphology and Morphometry: Is It a Prerequisite for Future Pathological Investigations? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1392:85-105. [DOI: 10.1007/978-3-031-13021-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|
13
|
Molecular Mechanisms Underlying Twin-to-Twin Transfusion Syndrome. Cells 2022; 11:cells11203268. [PMID: 36291133 PMCID: PMC9600593 DOI: 10.3390/cells11203268] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 11/17/2022] Open
Abstract
Twin-to-twin transfusion syndrome is a unique disease and a serious complication occurring in 10–15% of monochorionic multiple pregnancies with various placental complications, including hypoxia, anemia, increased oxidative stress, and ischemia-reperfusion injury. Fetoscopic laser photocoagulation, a minimally invasive surgical procedure, seals the placental vascular anastomoses between twins and dramatically improves the survival rates in twin-to-twin transfusion syndrome. However, fetal demise still occurs, suggesting the presence of causes other than placental vascular anastomoses. Placental insufficiency is considered as the main cause of fetal demise in such cases; however, little is known about its underlying molecular mechanisms. Indeed, the further association of the pathogenic mechanisms involved in twin-to-twin transfusion syndrome placenta with several molecules and pathways, such as vascular endothelial growth factor and the renin–angiotensin system, makes it difficult to understand the underlying pathological conditions. Currently, there are no effective strategies focusing on these mechanisms in clinical practice. Certain types of cell death due to oxidative stress might be occurring in the placenta, and elucidation of the molecular mechanism underlying this cell death can help manage and prevent it. This review reports on the molecular mechanisms underlying the development of twin-to-twin transfusion syndrome for effective management and prevention of fetal demise after fetoscopic laser photocoagulation.
Collapse
|
14
|
Xu N, Zhou X, Shi W, Ye M, Cao X, Chen S, Xu C. Integrative analysis of circulating microRNAs and the placental transcriptome in recurrent pregnancy loss. Front Physiol 2022; 13:893744. [PMID: 35991164 PMCID: PMC9390878 DOI: 10.3389/fphys.2022.893744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Recurrent pregnancy loss (RPL) is a major type of pathological pregnancy that still lacks reliable early diagnosis and effective treatment. The placenta is critical to fetal development and pregnancy success because it participates in critical processes such as early embryo implantation, vascular remodeling, and immunological tolerance. RPL is associated with abnormalities in the biological behavior of placental villous trophoblasts, resulting in aberrant placental function. MicroRNAs (miRNAs) are increasingly being recognized as essential regulators of placental development, as well as potential biomarkers. In this study, plasma miRNAs and placental messenger RNAs (mRNAs) from RPL patients and normal pregnant (NP) controls were sequenced and analyzed. Compared to those in NP controls, 108 circulating miRNAs and 1199 placental mRNAs were differentially expressed in RPL samples. A total of 140 overlapping genes (overlapping between plasma miRNA target genes and actual placental disorder genes) were identified, and functional enrichment analysis showed that these genes were mainly related to cell proliferation, angiogenesis, and cell migration. The regulatory network among miRNAs, overlapping genes, and downstream biological processes was analyzed by protein–protein interactions and Cytoscape. Moreover, enriched mRNAs, which were predictive targets of the differentially expressed plasma miRNAs miR-766-5p, miR-1285-3p, and miR-520a-3p, were accordingly altered in the placenta. These results suggest that circulating miRNAs may be involved in the pathogenesis of RPL and are potential noninvasive biomarkers for RPL.
Collapse
Affiliation(s)
- Naixin Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xuanyou Zhou
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Weihui Shi
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Mujin Ye
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xianling Cao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Songchang Chen
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- *Correspondence: Songchang Chen, ; Chenming Xu,
| | - Chenming Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- *Correspondence: Songchang Chen, ; Chenming Xu,
| |
Collapse
|
15
|
Liu T, Wen L, Huang S, Han TL, Zhang L, Fu H, Li J, Tong C, Qi H, Saffery R, Baker PN, Kilby MD. Comprehensive Metabolomic Profiling of Cord Blood and Placental Tissue in Surviving Monochorionic Twins Complicated by Twin-Twin Transfusion Syndrome With or Without Fetoscopic Laser Coagulation Surgery: A Retrospective Cohort Study. Front Bioeng Biotechnol 2022; 10:786755. [PMID: 35528207 PMCID: PMC9070302 DOI: 10.3389/fbioe.2022.786755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
Objectives: To investigate metabolomic perturbations caused by twin-twin transfusion syndrome, metabolic changes associated with fetoscopic laser coagulation in both placental tissue and cord plasma, and to investigate differential metabolites pertinent to varying fetal outcomes, including hemodynamic status, birth weight, and cardiac function, of live-born babies. Methods: Placental tissue and cord plasma samples from normal term or uncomplicated preterm-born monochorionic twins and those complicated by twin-twin transfusion syndrome treated with or without fetoscopic laser coagulation were analyzed by high-performance liquid chromatography metabolomic profiling. Sixteen comparisons of different co-twin groups were performed. Partial least squares–discriminant analysis, metabolic pathway analysis, biomarker analysis, and Spearman’s correlation analysis were conducted based on differential metabolites used to determine potential biomarkers in different comparisons and metabolites that are pertinent to neonatal birth weight and left ventricular ejection fraction. Results: These metabolomic investigations showed that the cord plasma metabolome has a better performance in discriminating fetuses among different hemodynamic groups than placental tissue. The metabolic alteration of twin-twin transfusion syndrome in these two types of samples centers on fatty acid and lipid metabolism. The fetoscopic laser coagulation procedure improves the metabolomic change brought by this syndrome, making the metabolomes of the treated group less distinguishable from those of the control and preterm birth groups. Certain compounds, especially lipids and lipid-like molecules, are noted to be potential biomarkers of this morbid disease and pertinent to neonatal birth weight and ejection fraction. Conclusions: Fetoscopic laser coagulation can ameliorate the metabolomic alteration caused by twin-twin transfusion syndrome in placental tissue and cord plasma, which are involved mainly in fatty acid and lipid-like molecule metabolism. Certain lipids and lipid-like molecules are helpful in differentiating co-twins of different hemodynamic statuses and are significantly correlated with neonatal birth weight or ejection fraction.
Collapse
Affiliation(s)
- Tianjiao Liu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Li Wen
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuai Huang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ting-li Han
- International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lan Zhang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huijia Fu
- Department of Reproduction Health and Infertility, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Junnan Li
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chao Tong
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing Medical University, Chongqing, China
- *Correspondence: Chao Tong, ; Hongbo Qi,
| | - Hongbo Qi
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Women and Children’s Health Center, Chongqing, China
- *Correspondence: Chao Tong, ; Hongbo Qi,
| | - Richard Saffery
- Cancer, Disease and Developmental Epigenetics, Murdoch Children’s Research Institute, Parkville, VIC, Australia
- Department of Pediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Philip N. Baker
- College of Life Sciences, University of Leicester, Leicester, United Kingdom
| | - Mark D. Kilby
- Institute of Metabolism and System Research, University of Birmingham, Birmingham, United Kingdom
- Fetal Medicine Centre, Birmingham Women’s and Children’s Foundation Trust, Birmingham, United Kingdom
| |
Collapse
|
16
|
Sikovanyecz J, Vincze M, Földesi I, Németh G, Kozinszky Z. Angiogenic factors measured in aspirated placental tissue between the 10 + 6 and 18 + 3 weeks of gestation. Reprod Biol 2021; 21:100572. [PMID: 34798517 DOI: 10.1016/j.repbio.2021.100572] [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: 03/08/2021] [Revised: 09/05/2021] [Accepted: 10/23/2021] [Indexed: 11/28/2022]
Abstract
This study was designed to determine the level of vascular endothelial growth factor-A (VEGF-A), basic fibroblast growth factor (bFGF) and endothelial nitric oxide synthase (eNOS) in chorionic villi during in first and second trimester, and their association with nuchal translucency (NT) measured by ultrasound. Seventy-five singleton healthy pregnancies with no detected congenital malformation were collected for NT measurements and chorionic villus sampling (CVS). Concentrations of angiogenic factors were assayed in chorionic villi sampled between 10 + 6 and 18 + 3 weeks of gestation. ENOS level was steady during this gestational period, while the concentrations of VEGF-A and bFGF significantly decreased. Placental concentrations of VEGF-A and bFGF correlated positively with each other (semi-partial correlation in multivariable linear regression (r): 0.90) and both correlated negatively with the eNOS level (r: -0.64 and r: -0.83, respectively). NT was positively correlated with eNOS concentration and negatively correlated with bFGF levels (r: 0.85 and r: -0.78, respectively). Inverse correlation was found between gestational age and VEGF-A and bFGF concentrations (r: -0.57 and r: 0.73, respectively). Alterations of angiogenic factors in chorionic villi might be an adjunct modality to NT and foetal growth as sonographic markers.
Collapse
Affiliation(s)
- János Sikovanyecz
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Albert Szent-Györgyi Medical and Pharmaceutical Centre, University of Szeged, Szeged, Hungary
| | - Márió Vincze
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Albert Szent-Györgyi Medical and Pharmaceutical Centre, University of Szeged, Szeged, Hungary
| | - Imre Földesi
- Department of Laboratory Medicine, Faculty of Medicine, Albert Szent-Györgyi Medical and Pharmaceutical Centre, University of Szeged, Szeged, Hungary
| | - Gábor Németh
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Albert Szent-Györgyi Medical and Pharmaceutical Centre, University of Szeged, Szeged, Hungary
| | - Zoltan Kozinszky
- Department of Obstetrics and Gynaecology, Danderyd Hospital, Stockholm, Sweden.
| |
Collapse
|
17
|
Ji ZS, Jiang H, Xie Y, Wei QP, Yin XF, Ye JH, Quan XZ, Lan YL, Zhao M, Tian XL, Zhang YJ, Yang XZ. Chemerin promotes the pathogenesis of preeclampsia by activating CMKLR1/p-Akt/CEBPɑ axis and inducing M1 macrophage polarization. Cell Biol Toxicol 2021; 38:611-628. [PMID: 34398343 DOI: 10.1007/s10565-021-09636-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/13/2021] [Indexed: 11/30/2022]
Abstract
A higher ratio of M1/M2 macrophages and an elevated chemerin level are both related to increased risk of preeclampsia. However, the crosstalk between these two events and their collective contribution to preeclampsia are not well understood. In this study, we assessed the impacts of chemerin chemokine-like receptor 1 (CMKLR1)/p-Akt/CEBPα axis in regulating macrophage polarization and mediating the pathogenic effects of chemerin on preeclampsia. We showed that chemerin, in a dose- and time-dependent manner, stimulated M1 macrophage polarization, inhibited macrophage-induced trophoblast invasion and migration, and suppressed macrophage-mediated angiogenesis. All these chemerin-induced phenotypes are essentially mediated by sequentially CMKLR1, Akt activation, and CEBPα. Mechanistically, CEBPα acted as a transcriptional activator for both IRF8 and chemerin. In vivo, chemerin aggravated preeclampsia, while α-NETA, an inhibitor for CMKLR1, significantly suppressed M1 macrophage polarization and alleviated preeclampsia. In summary, chemerin, by activating CMKLR1/Akt/CEBPα axis, forms a positive feedback loop, promotes M1 macrophage polarization, suppresses trophoblast migration/invasion and angiogenesis, and contributes to preeclampsia. Therefore, targeting chemerin signaling may benefit the prevention and/or treatment of preeclampsia.
Collapse
Affiliation(s)
- Zhi-Song Ji
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Hua Jiang
- Department of Clinical Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Yue Xie
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Qi-Peng Wei
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Xiao-Fang Yin
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Jin-Hai Ye
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Xiao-Zhen Quan
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Yan-Li Lan
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Meng Zhao
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Xiao-Long Tian
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Ya-Jun Zhang
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Xue-Zhou Yang
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China.
| |
Collapse
|
18
|
Jaiman S, Romero R, Pacora P, Erez O, Jung E, Tarca AL, Bhatti G, Yeo L, Kim YM, Kim CJ, Kim JS, Qureshi F, Jacques SM, Gomez-Lopez N, Hsu CD. Disorders of placental villous maturation are present in one-third of cases with spontaneous preterm labor. J Perinat Med 2021; 49:412-430. [PMID: 33554577 PMCID: PMC8324068 DOI: 10.1515/jpm-2020-0138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 11/09/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Spontaneous preterm labor is an obstetrical syndrome accounting for approximately 65-70% of preterm births, the latter being the most frequent cause of neonatal death and the second most frequent cause of death in children less than five years of age worldwide. The purpose of this study was to determine and compare to uncomplicated pregnancies (1) the frequency of placental disorders of villous maturation in spontaneous preterm labor; (2) the frequency of other placental morphologic characteristics associated with the preterm labor syndrome; and (3) the distribution of these lesions according to gestational age at delivery and their severity. METHODS A case-control study of singleton pregnant women was conducted that included (1) uncomplicated pregnancies (controls, n=944) and (2) pregnancies with spontaneous preterm labor (cases, n=438). All placentas underwent histopathologic examination. Patients with chronic maternal diseases (e.g., chronic hypertension, diabetes mellitus, renal disease, thyroid disease, asthma, autoimmune disease, and coagulopathies), fetal malformations, chromosomal abnormalities, multifetal gestation, preeclampsia, eclampsia, preterm prelabor rupture of the fetal membranes, gestational hypertension, gestational diabetes mellitus, and HELLP (hemolysis, elevated liver enzymes and low platelet count) syndrome were excluded from the study. RESULTS Compared to the controls, the most prevalent placental lesions among the cases were the disorders of villous maturation (31.8% [106/333] including delayed villous maturation 18.6% [62/333] vs. 1.4% [6/442], q<0.0001, prevalence ratio 13.7; and accelerated villous maturation 13.2% [44/333] vs. 0% [0/442], q<0.001). Other lesions in decreasing order of prevalence included hypercapillarized villi (15.6% [68/435] vs. 3.5% [33/938], q<0.001, prevalence ratio 4.4); nucleated red blood cells (1.1% [5/437] vs. 0% [0/938], q<0.01); chronic inflammatory lesions (47.9% [210/438] vs. 29.9% [282/944], q<0.0001, prevalence ratio 1.6); fetal inflammatory response (30.1% [132/438] vs. 23.2% [219/944], q<0.05, prevalence ratio 1.3); maternal inflammatory response (45.5% [195/438] vs. 36.1% [341/944], q<0.01, prevalence ratio 1.2); and maternal vascular malperfusion (44.5% [195/438] vs. 35.7% [337/944], q<0.01, prevalence ratio 1.2). Accelerated villous maturation did not show gestational age-dependent association with any other placental lesion while delayed villous maturation showed a gestational age-dependent association with acute placental inflammation (q-value=0.005). CONCLUSIONS Disorders of villous maturation are present in nearly one-third of the cases of spontaneous preterm labor.
Collapse
Affiliation(s)
- Sunil Jaiman
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women's Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, Florida, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yeon Mee Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Chong Jai Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jung-Sun Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Faisal Qureshi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women's Hospital, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Pathology, Harper University Hospital, Detroit, Michigan, USA
| | - Suzanne M. Jacques
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women's Hospital, Wayne State University School of Medicine, Detroit, MI, USA
- Department of Pathology, Harper University Hospital, Detroit, Michigan, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
19
|
Anh ND, Thuong PH, Sim NT, Thao TTP, Anh LTL, Canh TTT, Dieu NV, Bac ND, Tong HV. Maternal vascular endothelial growth factor receptor and interleukin levels in pregnant women with twin-twin transfusion syndrome. Int J Med Sci 2021; 18:3206-3213. [PMID: 34400890 PMCID: PMC8364448 DOI: 10.7150/ijms.61014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/21/2021] [Indexed: 11/30/2022] Open
Abstract
Twin-twin transfusion syndrome (TTTS) is an unusual and serious condition that occurs in twin pregnancies when identical twins share a placenta but develop discordant amniotic fluid volumes. TTTS is associated with an increased risk of fetal death and birth defects if untreated. This study investigated the soluble levels of biomarkers including growth factors and interleukins in pregnant women with and without TTTS during pregnancy. We quantified plasma levels of VEGF-R1, VEGF-R2, IL-1β, IL-6 and IL-8 in twin pregnant women with (n=53) and without TTTS (n=72) and in women with single pregnancy (n=30) by ELISA and analyzed the association of maternal circulating biomarker levels with TTTS. Our results showed that maternal VEGF-R1 levels were significantly higher in twins compared to single pregnancy (P<0.05) and were decreased in the second trimester compared to the first trimester (P = 0.065, 0.019 and 0.072 for twins with and without TTTS and single pregnancy, respectively). VEGF-R2 levels had a trend to be lower in twins compared to single pregnancy. In addition, soluble VEGF-R1 and VEGF-R2 levels were significantly decreased while IL-6 levels were increased after surgical treatment with laser in twin pregnant women with TTTS (P = 0.016, 0.041 and 0.04, respectively). These results suggest that IL-6, VEGF-R1 and VEGF-R2 are involved in vascular regulation and stabilization in twin pregnancies and may contribute to the pathogenesis of TTTS and thus play a prognostic role in the surgical treatment of TTTS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nguyen Van Dieu
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University
| | - Nguyen Duy Bac
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University
| | - Hoang Van Tong
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University
| |
Collapse
|
20
|
PlGF Immunological Impact during Pregnancy. Int J Mol Sci 2020; 21:ijms21228714. [PMID: 33218096 PMCID: PMC7698813 DOI: 10.3390/ijms21228714] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022] Open
Abstract
During pregnancy, the mother’s immune system has to tolerate the persistence of paternal alloantigens without affecting the anti-infectious immune response. Consequently, several mechanisms aimed at preventing allograft rejection, occur during a pregnancy. In fact, the early stages of pregnancy are characterized by the correct balance between inflammation and immune tolerance, in which proinflammatory cytokines contribute to both the remodeling of tissues and to neo-angiogenesis, thus, favoring the correct embryo implantation. In addition to the creation of a microenvironment able to support both immunological privilege and angiogenesis, the trophoblast invades normal tissues by sharing the same behavior of invasive tumors. Next, the activation of an immunosuppressive phase, characterized by an increase in the number of regulatory T (Treg) cells prevents excessive inflammation and avoids fetal immuno-mediated rejection. When these changes do not occur or occur incompletely, early pregnancy failure follows. All these events are characterized by an increase in different growth factors and cytokines, among which one of the most important is the angiogenic growth factor, namely placental growth factor (PlGF). PlGF is initially isolated from the human placenta. It is upregulated during both pregnancy and inflammation. In this review, we summarize current knowledge on the immunomodulatory effects of PlGF during pregnancy, warranting that both innate and adaptive immune cells properly support the early events of implantation and placental development. Furthermore, we highlight how an alteration of the immune response, associated with PlGF imbalance, can induce a hypertensive state and lead to the pre-eclampsia (PE).
Collapse
|
21
|
Rossi C, Lees M, Mehta V, Heikura T, Martin J, Zachary I, Spencer R, Peebles DM, Shaw R, Karhinen M, Yla-Herttuala S, David AL. Comparison of Efficiency and Function of Vascular Endothelial Growth Factor Adenovirus Vectors in Endothelial Cells for Gene Therapy of Placental Insufficiency. Hum Gene Ther 2020; 31:1190-1202. [PMID: 32988220 PMCID: PMC7698978 DOI: 10.1089/hum.2020.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022] Open
Abstract
Severe fetal growth restriction (FGR) affects 1:500 pregnancies, is untreatable and causes serious neonatal morbidity and death. Reduced uterine blood flow (UBF) and lack of bioavailable VEGF due to placental insufficiency is a major cause. Transduction of uterine arteries in normal or FGR sheep and guinea pigs using an adenovirus (Ad) encoding VEGF isoforms A (Ad.VEGF-A165) and a FLAG-tagged pre-processed short form D (DΔNΔC, Ad.VEGF-DΔNΔC-FLAG) increases endothelial nitric oxide expression, enhances relaxation and reduces constriction of the uterine arteries and their branches. UBF and angiogenesis are increased long term, improving fetal growth in utero. For clinical trial development we compared Ad.VEGF vector transduction efficiency and function in endothelial cells (ECs) derived from different species. We aimed to compare the transduction efficiency and function of the pre-clinical study Ad. constructs (Ad.VEGF-A165, Ad.VEGF-DΔNΔC-FLAG) with the intended clinical trial construct (Ad.VEGF-DΔNΔC) where the FLAG tag is removed. We infected ECs from human umbilical vein, pregnant sheep uterine artery, pregnant guinea pig aorta and non-pregnant rabbit aorta, with increasing multiplicity of infection (MOI) for 24 or 48 hours of three Ad.VEGF vectors, compared to control Ad. containing the LacZ gene (Ad.LacZ). VEGF supernatant expression was analysed by ELISA. Functional assessment used tube formation assay and Erk-Akt phosphorylation by ELISA. VEGF expression was higher after Ad.VEGF-DΔNΔC-FLAG and Ad.VEGF-DΔNΔC transduction compared to Ad.VEGF-A165 in all EC types (*p < 0.001). Tube formation was higher in ECs transduced with Ad.VEGF-DΔNΔC in all species compared to other constructs (***p < 0.001, *p < 0.05 with rabbit aortic ECs). Phospho-Erk and phospho-Akt assays displayed no differences between the three vector constructs, whose effect was, as in other experiments, higher than Ad.LacZ (***p < 0.001). In conclusion, we observed high transduction efficiency and functional effects of Ad.VEGF-DΔNΔC vector with comparability in major pathway activation to constructs used in pre-clinical studies, supporting its use in a clinical trial.
Collapse
Affiliation(s)
- Carlo Rossi
- Elizabeth Garrett Anderson Institute for Women's Health
- Centre for Cardiovascular Biology and Medicine; University College London, London, United Kingdom
| | - Mark Lees
- Elizabeth Garrett Anderson Institute for Women's Health
- Centre for Cardiovascular Biology and Medicine; University College London, London, United Kingdom
| | - Vedanta Mehta
- Centre for Cardiovascular Biology and Medicine; University College London, London, United Kingdom
| | - Tommi Heikura
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - John Martin
- Centre for Cardiovascular Biology and Medicine; University College London, London, United Kingdom
| | - Ian Zachary
- Centre for Cardiovascular Biology and Medicine; University College London, London, United Kingdom
| | | | | | | | | | - Seppo Yla-Herttuala
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - Anna L. David
- Elizabeth Garrett Anderson Institute for Women's Health
| |
Collapse
|
22
|
Sahay A, Kale A, Joshi S. Role of neurotrophins in pregnancy and offspring brain development. Neuropeptides 2020; 83:102075. [PMID: 32778339 DOI: 10.1016/j.npep.2020.102075] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 02/06/2023]
Abstract
Neurotrophins are a family of functionally and structurally related proteins which play a key role in the survival, development, and function of neurons in both the central and peripheral nervous systems. Brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4) are the family members of neurotrophins. Neurotrophins play a crucial role in influencing the development of the brain and learning and memory processes. Studies demonstrate that they also play crucial role in influencing reproductive and immune systems. Neurotrophins have been shown to influence various processes in the mother, placenta, and fetus during pregnancy. Development and maturation of feto-placental unit and the fetal growth trajectories are influenced by neurotrophins. In addition to neurotrophins, neuropeptides like neuropeptide Y also play a crucial role during various processes of pregnancy and during fetal brain development. Neurotrophins have also been shown to have a cross talk with various angiogenic factors and influence placental development. Alterations in the levels of neurotrophins and neuropeptides lead to placental pathologies resulting in various pregnancy complications like preeclampsia, intrauterine growth restriction and preterm births. Studies in animals have reported low levels of maternal micronutrients like folic acid, vitamin B12 and omega-3 fatty acids influence brain neurotrophins resulting in impaired cognitive functioning in the offspring. Maternal nutrition is also known to affect the expression of neuropeptides. It is essential to understand the role of various neurotrophins across various stages of pregnancy and its relationship with neurodevelopmental outcomes in children. This will lead to early prediction of poor neurodevelopmental outcomes. The present review describes evidence describing the role of neurotrophins in determining pregnancy outcome and altered neurodevelopment in the offspring. The possible mechanism through which maternal nutrition influences neurotrophins and neuropeptides to regulate offspring brain development and function is also discussed.
Collapse
Affiliation(s)
- Akriti Sahay
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Anvita Kale
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Sadhana Joshi
- Mother and Child Health, Interactive Research School for Health Affairs, Bharati Vidyapeeth (Deemed to be University), Pune, India.
| |
Collapse
|
23
|
Jaiman S, Romero R, Pacora P, Jung E, Bhatti G, Yeo L, Kim YM, Kim B, Kim CJ, Kim JS, Qureshi F, Jacques SM, Erez O, Gomez-Lopez N, Hsu CD. Disorders of placental villous maturation in fetal death. J Perinat Med 2020; 0:/j/jpme.ahead-of-print/jpm-2020-0030/jpm-2020-0030.xml. [PMID: 32238609 PMCID: PMC8262362 DOI: 10.1515/jpm-2020-0030] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 02/21/2020] [Indexed: 12/22/2022]
Abstract
Objective The aims of this study were to ascertain the frequency of disorders of villous maturation in fetal death and to also delineate other placental histopathologic lesions in fetal death. Methods This was a retrospective observational cohort study of fetal deaths occurring among women between January 2004 and January 2016 at Hutzel Women's Hospital, Detroit, MI, USA. Cases comprised fetuses with death beyond 20 weeks' gestation. Fetal deaths with congenital anomalies and multiple gestations were excluded. Controls included pregnant women without medical/obstetrical complications and delivered singleton, term (37-42 weeks) neonate with 5-min Apgar score ≥7 and birthweight between the 10th and 90th percentiles. Results Ninety-two percent (132/143) of placentas with fetal death showed placental histologic lesions. Fetal deaths were associated with (1) higher frequency of disorders of villous maturation [44.0% (64/143) vs. 1.0% (4/405), P < 0.0001, prevalence ratio, 44.6; delayed villous maturation, 22% (31/143); accelerated villous maturation, 20% (28/143); and maturation arrest, 4% (5/143)]; (2) higher frequency of maternal vascular malperfusion lesions [75.5% (108/143) vs. 35.7% (337/944), P < 0.0001, prevalence ratio, 2.1] and fetal vascular malperfusion lesions [88.1% (126/143) vs. 19.7% (186/944), P < 0.0001, prevalence ratio, 4.5]; (3) higher frequency of placental histologic patterns suggestive of hypoxia [59.0% (85/143) vs. 9.3% (82/942), P < 0.0001, prevalence ratio, 6.8]; and (4) higher frequency of chronic inflammatory lesions [53.1% (76/143) vs. 29.9% (282/944), P < 0.001, prevalence ratio 1.8]. Conclusion This study demonstrates that placentas of women with fetal death were 44 times more likely to present disorders of villous maturation compared to placentas of those with normal pregnancy. This suggests that the burden of placental disorders of villous maturation lesions is substantial.
Collapse
Affiliation(s)
- Sunil Jaiman
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, Florida, USA
| | - Percy Pacora
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Gaurav Bhatti
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Lami Yeo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yeon Mee Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Bomi Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Chong Jai Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jung-Sun Kim
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Faisal Qureshi
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| | - Suzanne M. Jacques
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Pathology, Hutzel Women’s Hospital, Wayne State University School of Medicine, Detroit, MI, USA
| | - Offer Erez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Soroka University Medical Center, School of Medicine, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Chaur-Dong Hsu
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
24
|
Kidron D, Bar-Lev Y, Tsarfaty I, Many A, Tauman R. The effect of maternal obstructive sleep apnea on the placenta. Sleep 2020; 42:5418772. [PMID: 30903184 DOI: 10.1093/sleep/zsz072] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 02/07/2019] [Accepted: 03/14/2019] [Indexed: 01/25/2023] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea (OSA) during pregnancy has been associated with adverse maternal outcomes. However, the effect of maternal OSA on fetal growth is less clear. The placenta is a critical organ for fetal growth and development and the principal determinant of birthweight. We aimed to investigate the effect of maternal OSA on placental growth and function. METHODS Placentas of women recruited to a prospective longitudinal study were consecutively obtained immediately after delivery. Each placenta was measured for length, width, and thickness. Total RNA was isolated for gene expression analysis of VEGF, VEGF receptor, PIGF, and leptin. Histological and morphometric evaluations of the placenta were performed. RESULTS A total of 53 placentas were investigated. Ten women (19%) had OSA, and the weight of their placentas was significantly higher compared with the placentas of the controls (526.1 ± 83.9 vs. 425.7 ± 95.5 g, p = 0.004). There was a significant positive correlation between placental weight and the log apnea-hypopnea index even after controlling for maternal body mass index (BMI; r = 0.31, p = 0.04). The birthweight/placental weight ratio was significantly lower in women with OSA compared with controls (p = 0.03). Placental weight and newborn triceps adiposity thickness correlated positively after controlling for maternal BMI (r = 0.29, p = 0.04). Leptin expression was 1.8-fold higher in placentas of women with OSA compared with controls (p = 0.02). No histological differences were found between the groups. CONCLUSIONS Maternal OSA is associated with increased placental weight that correlated with OSA severity and neonatal adiposity independently of maternal BMI. Placental leptin overexpression may mediate/underlie the above findings.Trial Registration: Clinical Trials NCT00931099.
Collapse
Affiliation(s)
- Debora Kidron
- The Pathology Department, Meir Medical Center, Kfar Saba.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yamit Bar-Lev
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Clinical Microbiology and Immunology, Lis Maternity Hospital, Tel Aviv, Israel
| | - Ilan Tsarfaty
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Clinical Microbiology and Immunology, Lis Maternity Hospital, Tel Aviv, Israel
| | - Ariel Many
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Obstetrics and Gynceology, Lis Maternity Hospital, Tel Aviv, Israel
| | - Riva Tauman
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.,The Sleep Disorders Center, Tel Aviv Medical Center, all affiliated to Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
25
|
Bhattacharjee D, Mondal SK, Garain P, Mandal P, Ray RN, Dey G. Histopathological study with immunohistochemical expression of vascular endothelial growth factor in placentas of hyperglycemic and diabetic women. J Lab Physicians 2020; 9:227-233. [PMID: 28966481 PMCID: PMC5607748 DOI: 10.4103/jlp.jlp_148_16] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
AIMS AND OBJECTIVES Spectrum of hyperglycemia in pregnancy includes gestational diabetes mellitus (GDM), mild hyperglycemia, and overt diabetes. Many authors have worked on morphological changes of the placenta in diabetes, but few studies have correlated histopathological changes with vascular endothelial growth factor (VEGF) immunoexpression. The aim of this study was to detect different histopathological changes in various groups of diabetic placentas and to correlate with VEGF immunoexpression. MATERIALS AND METHODS Pregnant women were screened for diabetes. They were subsequently divided into normoglycemic (12 cases), GDM (33 cases), mild hyperglycemic (13 cases), and overt diabetes (18 cases). Placentas collected were subjected to histopathological examination. VEGF expressions were studied by immunohistochemistry. RESULTS Overt diabetic placenta displayed villous immaturity (44.4%), villous edema (38.9%), chorangiosis (61.1%), fibrinoid substance deposition (38.9%), and Hofbauer cell hyperplasia in 44.4% cases. GDM placentas displayed villous immaturity (45.5%), villous edema (45.5%), chorangiosis (42.4%), and fibrinoid substance deposition in 75.6% cases. Mild hyperglycemic placentas displayed villous immaturity (38.5%), chorangiosis (61.5%), and fibrinoid substance deposition in 61.5% cases. VEGF immunoexpression in GDM placentas was absent in all placental components except syncytiotrophoblast. VEGF expression in overt diabetic placentas was increased in syncytiotrophoblast and capillary endothelium compared to normoglycemic placentas. Mild hyperglycemic placentas expressed similar VEGF expression in all components when compared to normoglycemic controls. However, it displayed weak expression in vessel endothelium. CONCLUSION Histopathological changes in diabetic placentas might be a consequence of altered or abnormal VEGF expression in diabetic placentas. Pathogenesis and VEGF expression in GDM placentas are significantly different from overt diabetic placentas.
Collapse
Affiliation(s)
| | - Santosh Kumar Mondal
- Department of Pathology, Bankura Sammilani Medical College, Bankura, West Bengal, India
| | - Pratima Garain
- Department of Gynaecology and Obstetrics, Bankura Sammilani Medical College, Bankura, West Bengal, India
| | - Palash Mandal
- Department of Pathology, School of Medicine, Sagore Dutta Hospital, Kolkata, West Bengal, India
| | - Rudra Narayan Ray
- Department of Pathology, Bankura Sammilani Medical College, Bankura, West Bengal, India
| | - Goutam Dey
- Department of Pathology, Bankura Sammilani Medical College, Bankura, West Bengal, India
| |
Collapse
|
26
|
Two patterns of cytokine production by placental macrophages. Placenta 2020; 91:1-10. [PMID: 31941612 DOI: 10.1016/j.placenta.2020.01.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/09/2019] [Accepted: 01/07/2020] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Macrophages participate in the regulation immune and morphogenetic events in the placenta. However, these roles remain unclear for placental macrophages (Hofbauer cells). The aims of this study were to characterize the consecutive steps of cytokine production (intracellular synthesis and secretion) in placental macrophages in early and late gestation and to compare the secretory profiles of placental macrophages and villous tissue. METHODS Macrophages and villous tissue were isolated from placentas obtained from normal pregnancies at either 9-12 or 38-40 weeks of gestation. Intracellular cytokines were determined by flow cytometry after staining with monoclonal antibodies. Secreted cytokines were quantified by cytometric bead array and ELISA. RESULTS Two patterns of cytokine production were revealed in placental macrophages. Cytokines in the first group (IL-1, IL-6, IL-8, IL-10, TNFα) demonstrated low basal production and were stimulated by bacterial endotoxin. Cytokines in the second group (IL-11, IL-17A, IL-17F, TGF-β, VEGF) were characterized by constitutive production and did not respond to stimulation. Gestational age-dependent changes were observed: basal secretion of TNFα and IL-8 increased whereas IL-11 and IL-17 secretion decreased in third-trimester macrophages compared with the first-trimester cells. Comparison of cytokine production at the cellular and tissue levels suggested the contribution of the placental macrophages both in intraplacental and extraplacental cytokine production. DISCUSSION It would be safe to assume that the two patterns of cytokine production, revealed in our study, correspond to two regulatory roles of placental macrophages: "immune" and "morphogenetic". The inflammatory phenotype of macrophages is attenuated in early gestation and increases with the progression of pregnancy. The cytokines of the first group supposedly contribute to both local and extraplacental levels, whereas the cytokine effects of the second group are more likely confined to the placental tissue.
Collapse
|
27
|
Depoix CL, Colson A, Hubinont C, Debieve F. Impaired vascular endothelial growth factor expression and secretion during in vitro differentiation of human primary term cytotrophoblasts. Angiogenesis 2020; 23:221-230. [PMID: 31894427 DOI: 10.1007/s10456-019-09702-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/11/2019] [Indexed: 01/05/2023]
Abstract
Vascular endothelial growth factor A (VEGF-A) is one of the main growth factors involved in placental vasculogenesis and angiogenesis, but its placental expression is still ambiguous. During in vitro cultures of primary term cytotrophoblasts, VEGF could not be detected in the supernatants by enzyme-linked immunosorbent assays (ELISA). One hypothesis is that VEGF is immediately and completely bound to its soluble receptor after secretion, and cannot be recognized by the antibodies used in the commercial ELISA kits. We decided to verify this hypothesis by measuring VEGF-A expression during in vitro cultures of primary term cytotrophoblasts. Term cytotrophoblasts were cultured under 21% and 2.5% O2 for 4 days. VEGF-A transcripts were quantified by real-time polymerase chain reaction. The proteins from cell lysates and concentrated media were separated by polyacrylamide gel electrophoresis (PAGE) under denaturing and reducing conditions, and VEGF-A immunodetected by western blotting. VEGF mRNA expression did not increase during in vitro cell differentiation under 21% O2, but slightly increased under 2.5% O2 only at 24 h. VEGF-A monomer was not detected in the cell lysates and in the concentrated supernatants, while a ~ 42 KDa band corresponding to the precursor L-VEGF was detected in all the cellular extracts. Isolated term villous cytotrophoblasts produce the L-VEGF precursor but they do not secrete VEGF-A even under low-oxygen tension. The question remains about the origin of VEGF in pregnancy but also about the biological role of L-VEGF, which can represent a form of storage for rapid VEGF secretion when needed.
Collapse
Affiliation(s)
- Christophe Louis Depoix
- Department of Obstetrics, Institut de Recherche Clinique Et Experimentale (IREC), Université Catholique de Louvain, Avenue Mounier 52, 5th floor, Woluwe-Saint-Lambert, 1200, Bruxelles, Belgium.
| | - Arthur Colson
- Department of Obstetrics, Institut de Recherche Clinique Et Experimentale (IREC), Université Catholique de Louvain, Avenue Mounier 52, 5th floor, Woluwe-Saint-Lambert, 1200, Bruxelles, Belgium
| | - Corinne Hubinont
- Department of Obstetrics, Institut de Recherche Clinique Et Experimentale (IREC), Université Catholique de Louvain, Avenue Mounier 52, 5th floor, Woluwe-Saint-Lambert, 1200, Bruxelles, Belgium
| | - Frederic Debieve
- Department of Obstetrics, Institut de Recherche Clinique Et Experimentale (IREC), Université Catholique de Louvain, Avenue Mounier 52, 5th floor, Woluwe-Saint-Lambert, 1200, Bruxelles, Belgium
| |
Collapse
|
28
|
Venkata Surekha M, Singh S, Sarada K, Sailaja G, Balakrishna N, Srinivas M, Uday Kumar P. Study on the Effect of Severity of Maternal Iron Deficiency Anemia on Regulators of Angiogenesis in Placenta. Fetal Pediatr Pathol 2019; 38:361-375. [PMID: 31130046 DOI: 10.1080/15513815.2019.1587120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Aims: In this study, we hypothesized that maternal anemia leads to altered expression of angiogenic proteins vascular endothelial growth factor (VEGF), placental growth factor (PLGF), nitrotyrosine (NT) residues, and endothelial nitric oxide synthase (e-NOS) in the placenta. Hence, we study the expression of the abovementioned proteins in the placentas of mothers with different grades of anemia. Materials and methods: Our study was conducted in 48 pregnant women (36-40 weeks of gestation), who were divided into four groups-normal, mild, moderate, and severe anemia. After delivery, the expression of the angiogenic proteins was studied in their placentas by immunohistochemistry. Results: In our study, 58.3% of the pregnant women were anemic, among which 20.83% had mild anemia, 18.75% had moderate anemia, and 18.75% had severe anemia. Immunohistochemical staining intensity for VEGF, PLGF, NT residues, and e-NOS proteins was observed to be higher in the placentas of anemic women when compared with the non-anemic women. Conclusion: Our study showed that there is an increased expression of angiogenic proteins in the placentas of anemic mothers, which probably is an adaptive response leading to changes in placental vessels.
Collapse
Affiliation(s)
| | - Sapna Singh
- National Institute of Nutrition, Pathology Division , Tarnaka , Hyderabad, Telangana , India
| | - Krishnakumar Sarada
- National Institute of Nutrition, Pathology Division , Tarnaka , Hyderabad, Telangana , India
| | - Gummadi Sailaja
- National Institute of Nutrition, Pathology Division , Tarnaka , Hyderabad, Telangana , India
| | - Nagalla Balakrishna
- National Institute of Nutrition, Biostatistics , Tarnaka , Hyderabad, Telangana , India
| | - Myadara Srinivas
- National Institute of Nutrition, NCLAS , Tarnaka , Hyderabad, Telangana , India
| | - Putcha Uday Kumar
- National Institute of Nutrition, Pathology and Microbiology , Tarnaka , Hyderabad, Telangana , India
| |
Collapse
|
29
|
Wang X, Sun T, Chen G, Gao H. Association between Vascular Endothelial Growth Factor Gene Polymorphisms and Pre-Eclampsia Susceptibility: An Updated Meta-Analysis. Immunol Invest 2019; 49:120-133. [PMID: 31455120 DOI: 10.1080/08820139.2019.1659812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Objectives: The aim of this study was to assess the genetic association between vascular endothelial growth factor (VEGF) gene polymorphisms and the risk of pre-eclampsia (PE).Methods: A systematic literature search of several databases (PubMed, Embase, and the China National Knowledge Infrastructure (CNKI)) was conducted for case-control trials comparing VEGF polymorphisms (+936C/T, -634G/C, -2578C/A, and -1154G/A) with the risk of PE. Meta-analysis was performed using the Stata 12.0 software.Results: Twenty-three case-control studies on a total of 2597 PE patients and 3075 controls were included in our meta-analysis. The +936C/T polymorphism was observed to be associated with the risk of PE in the overall population (T vs. C: odds ratios (OR) = 1.434, 95% confidence interval (CI) = 1.120-1.836, P = .004). However, the -634G/C, -2578C/A, and -1154G/A polymorphisms showed no association with the risk of PE. A subgroup analysis based on ethnicity found that the +936C/T polymorphism was associated with the risk of PE in both Europeans and Asians. Furthermore, the -634G/C polymorphism was found to be associated with the risk of PE in Europeans (C vs. G: OR = 1.428, 95% CI = 1.141-1.778, P = .002). The polymorphisms at other loci were not associated with the risk of PE.Conclusion: This meta-analysis suggests that VEGF +936C/T polymorphism, rather than -634G/C, -2578C/A, or -1154G/A polymorphisms, is associated with the risk of PE in the overall study population. However, the -634G/C polymorphism may be associated with the risk of developing PE in Europeans.
Collapse
Affiliation(s)
- Xiaoyi Wang
- Internal Medicine Department, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Tao Sun
- Internal Medicine Department, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Guochang Chen
- Internal Medicine Department, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Hong Gao
- Internal Medicine Department, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Yao Y, Xu XH, Jin L. Macrophage Polarization in Physiological and Pathological Pregnancy. Front Immunol 2019; 10:792. [PMID: 31037072 PMCID: PMC6476302 DOI: 10.3389/fimmu.2019.00792] [Citation(s) in RCA: 431] [Impact Index Per Article: 86.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Abstract
The immunology of pregnancy is complex and poorly defined. During the complex process of pregnancy, macrophages secrete many cytokines/chemokines and play pivotal roles in the maintenance of maternal-fetal tolerance. Here, we summarized the current knowledge of macrophage polarization and the mechanisms involved in physiological or pathological pregnancy processes, including miscarriage, preeclampsia, and preterm birth. Although current evidence provides a compelling argument that macrophages are important in pregnancy, our understanding of the roles and mechanisms of macrophages in pregnancy is still rudimentary. Since macrophages exhibit functional plasticity, they may be ideal targets for therapeutic manipulation during pathological pregnancy. Additional studies are needed to better define the functions and mechanisms of various macrophage subsets in both normal and pathological pregnancy.
Collapse
Affiliation(s)
- Yongli Yao
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiang-Hong Xu
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liping Jin
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Laakkonen JP, Lähteenvuo J, Jauhiainen S, Heikura T, Ylä-Herttuala S. Beyond endothelial cells: Vascular endothelial growth factors in heart, vascular anomalies and placenta. Vascul Pharmacol 2018; 112:91-101. [PMID: 30342234 DOI: 10.1016/j.vph.2018.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 10/16/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022]
Abstract
Vascular endothelial growth factors regulate vascular and lymphatic growth. Dysregulation of VEGF signaling is connected to many pathological states, including hemangiomas, arteriovenous malformations and placental abnormalities. In heart, VEGF gene transfer induces myocardial angiogenesis. Besides vascular and lymphatic endothelial cells, VEGFs affect multiple other cell types. Understanding VEGF biology and its paracrine signaling properties will offer new targets for novel treatments of several diseases.
Collapse
Affiliation(s)
- Johanna P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| | - Johanna Lähteenvuo
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Suvi Jauhiainen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tommi Heikura
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland; Science Service Center, Kuopio University Hospital, Kuopio, Finland; Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
32
|
Keshavarzi F, Shahrakipoor M, Teimoori B, Yaghmaei M, Narooei-Nejad M, Rasooli A, Salimi S. Association of the placental VEGF promoter polymorphisms and VEGF mRNA expression with preeclampsia. Clin Exp Hypertens 2018; 41:274-279. [DOI: 10.1080/10641963.2018.1469644] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Affiliation(s)
- Farshid Keshavarzi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, zahedan, Iran (the Islamic Republic of)
| | - Mahnaz Shahrakipoor
- Department of Biostatistics and Epidemiology, School of Public Health, Zahedan University of Medical Sciences, zahedan, Iran (the Islamic Republic of)
| | - Batool Teimoori
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran (the Islamic Republic of)
| | - Minoo Yaghmaei
- Department of Obstetrics and Gynecology, School of Medicine, Shahid Beheshty University of Medical Sciences, tehran, Iran (the Islamic Republic of)
| | - Mehrnaz Narooei-Nejad
- Department of Genetics, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran (the Islamic Republic of)
| | - Ava Rasooli
- Department of Laboratory Science, Paramedical School, zahedan, Iran (the Islamic Republic of)
| | - Saeedeh Salimi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, zahedan, Iran (the Islamic Republic of)
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran (the Islamic Republic of)
| |
Collapse
|
33
|
Alahakoon TI, Zhang W, Arbuckle S, Zhang K, Lee V. Reduced angiogenic factor expression in intrauterine fetal growth restriction using semiquantitative immunohistochemistry and digital image analysis. J Obstet Gynaecol Res 2018; 44:861-872. [DOI: 10.1111/jog.13592] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/23/2017] [Indexed: 11/26/2022]
Affiliation(s)
- Thushari I. Alahakoon
- The University of Sydney, Westmead Clinical School; Sydney New South Wales Australia
- Westmead Institute for Maternal and Fetal Medicine; Westmead Hospital; Sydney New South Wales Australia
| | - Weiyi Zhang
- Westmead Institute for Maternal and Fetal Medicine; Westmead Hospital; Sydney New South Wales Australia
| | - Susan Arbuckle
- Anatomical Pathology; The Children's Hospital; Sydney New South Wales Australia
| | - Kewei Zhang
- The University of Sydney, Westmead Clinical School; Sydney New South Wales Australia
- Westmead Institute for Maternal and Fetal Medicine; Westmead Hospital; Sydney New South Wales Australia
| | - Vincent Lee
- The University of Sydney, Westmead Clinical School; Sydney New South Wales Australia
- Renal Medicine; Westmead Hospital; Sydney New South Wales Australia
| |
Collapse
|
34
|
Brodowski L, Burlakov J, Hass S, von Kaisenberg C, von Versen-Höynck F. Impaired functional capacity of fetal endothelial cells in preeclampsia. PLoS One 2017; 12:e0178340. [PMID: 28542561 PMCID: PMC5441640 DOI: 10.1371/journal.pone.0178340] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/11/2017] [Indexed: 12/02/2022] Open
Abstract
Objectives Preeclampsia is one of the main contributers to maternal and fetal morbidity and mortality during pregnancy. A history of preeclampsia puts mother and offspring at an increased cardiovascular risk in later life. We hypothesized that at the time of birth functional impairments of fetal endothelial cells can be detected in pregnancies complicated by preeclampsia and that a therapeutic intervention using 1,25 (OH)2 vitamin D3 can reverse the adverse effects of preeclampsia on cell function. Methods Human umbilical vein endothelial cells (HUVEC) were isolated from umbilical cords obtained from preeclamptic (N = 12) and uncomplicated pregnancies (N = 13, control). Placental villous tissue fragments from uncomplicated term pregnancies were incubated in explant culture for 48 h at 2% (hypoxia), 8% or 21% O2. Explant conditioned media (CM) was collected and pooled according to oxygen level. We compared the ability of preeclampsia vs. control HUVEC to migrate, proliferate, and form tubule-like networks in a Matrigel assay, in the presence/absence of CM and 1,25(OH)2 vitamin D3. Results HUVEC from preeclamptic pregnancies showed reduced migration (P = 0.04) and tubule formation (P = 0.04), but no change in proliferation (P = 0.16) compared to healthy pregnancies. Placental villous explant CM derived from 2% O2 incubations significantly reduced HUVEC migration, when compared to non-CM (P = 0.04). Vitamin D3 improved HUVEC function in neither of the groups. There was no significant difference in VEGF gene expression between healthy and preeclamptic pregnancies and no effect of Vitamin D3 on VEGF expression. Conclusions Reduced functional abilities of fetal endothelial cells from preeclamptic pregnancies suggests that disease pathways, possibly originating from the dysfunctional placenta, negatively impact fetal endothelium. The neutral effect of 1,25(OH)2 vitamin D3 contrasts with previous findings that vitamin D rescues the poor migration, proliferation and tubule formation exhibited by cord blood fetal endothelial progenitor cells from preeclamptic pregnancies. Further investigations to distinguish pathways by which offspring exposed to preeclampsia are at risk for cardiovascular disease are needed.
Collapse
Affiliation(s)
- Lars Brodowski
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Jennifer Burlakov
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | - Sarah Hass
- Department of Obstetrics and Gynecology, Hannover Medical School, Hannover, Germany
| | | | | |
Collapse
|
35
|
Chon AH, Chavira ER, Wilson ML, Ingles SA, Llanes A, Chmait RH. The impact of laser surgery on angiogenic and anti-angiogenic factors in twin–twin transfusion syndrome: a prospective study. J Matern Fetal Neonatal Med 2017; 31:1085-1091. [DOI: 10.1080/14767058.2017.1309020] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Andrew H. Chon
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Emiliano R. Chavira
- Department of Maternal-Fetal Medicine, San Gabriel Valley Perinatal Medical Group, Monterey Park, CA, USA
| | - Melissa L. Wilson
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sue A. Ingles
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arlyn Llanes
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ramen H. Chmait
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
36
|
Li S, Roberson MS. Dlx3 and GCM-1 functionally coordinate the regulation of placental growth factor in human trophoblast-derived cells. J Cell Physiol 2017; 232:2900-2914. [PMID: 27996093 DOI: 10.1002/jcp.25752] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 11/08/2022]
Abstract
Placental growth factor (PGF) is abundantly expressed by trophoblast cells within human placentae and is important for trophoblast development and placental vascularization. Circulating maternal serum levels of PGF are dynamically upregulated across gestation in normal pregnancies, whereas low circulating levels and placental production of PGF have been implicated in the pathogenesis of preeclampsia and other gestational diseases. However, the underlying molecular mechanism of regulating PGF expression in the human placenta remains poorly understood. In this study, we demonstrated that transcription factors Distal-less 3 (DLX3) and Glial cell missing-1 (GCM1) were both sufficient and required for PGF expression in human trophoblast-derived cells by overexpression and knockdown approaches. Surprisingly, while DLX3 and GCM1 were both positive regulators of PGF, co-overexpression of DLX3 and GCM1 led to an antagonist effect on PGF expression on the endogenous gene and a luciferase reporter. Further, deletion and site-directed mutagenesis studies identified a novel regulatory element on the PGF promoter mediating both DLX3- and GCM1-dependent PGF expression. This regulatory region was also found to be essential for the basal activity of the PGF promoter. Finally, Chromatin-immunoprecipitation (ChIP) assays revealed colocalization of DLX3 and GCM1 at the identified regulatory region on the PGF promoter. Taken together, our studies provide important insights into intrinsic regulation of human placental PGF expression through the functional coordination of DLX3 and GCM1, and are likely to further the understanding of pathogenesis of PGF dysregulation in preeclampsia and other disease conditions.
Collapse
Affiliation(s)
- Sha Li
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| | - Mark S Roberson
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York
| |
Collapse
|
37
|
Hoeller A, Ehrlich L, Golic M, Herse F, Perschel FH, Siwetz M, Henrich W, Dechend R, Huppertz B, Verlohren S. Placental expression of sFlt-1 and PlGF in early preeclampsia vs. early IUGR vs. age-matched healthy pregnancies. Hypertens Pregnancy 2017; 36:151-160. [PMID: 28609172 DOI: 10.1080/10641955.2016.1273363] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To investigate whether differences between early preeclampsia and early fetal growth restriction can be explained by differential placental expression patterns of sFlt-1, Flt-1, and PlGF. METHODS Placental tissues and maternal blood samples from six cases of preeclampsia, seven IUGR, and six age-matched controls were studied for mRNA and protein levels as well as protein localization and expression intensity. RESULTS Neither placental PlGF mRNA and protein expression nor placental villous trophoblast expression intensity of PlGF was altered by placental dysfunction. CONCLUSION High sFlt-1 concentrations may account for diminished maternal serum PlGF levels.
Collapse
Affiliation(s)
- Alice Hoeller
- a Department of Obstetrics , Charité University Medicine , Berlin , Germany
| | - Laura Ehrlich
- a Department of Obstetrics , Charité University Medicine , Berlin , Germany
| | - Michaela Golic
- a Department of Obstetrics , Charité University Medicine , Berlin , Germany.,b HELIOS Clinic, Experimental and Clinical Research Center, Max-Delbrueck Center , Berlin-Buch , Germany
| | - Florian Herse
- b HELIOS Clinic, Experimental and Clinical Research Center, Max-Delbrueck Center , Berlin-Buch , Germany
| | - Frank H Perschel
- c Laboratory Medicine and Toxicology , Charité University Medicine Berlin und Labor Berlin - Charité, Vivantes GmbH , Berlin , Germany
| | - Monika Siwetz
- d Institute of Cell Biology, Histology and Embryology, Medical University of Graz , Graz , Austria
| | - Wolfgang Henrich
- a Department of Obstetrics , Charité University Medicine , Berlin , Germany
| | - Ralf Dechend
- b HELIOS Clinic, Experimental and Clinical Research Center, Max-Delbrueck Center , Berlin-Buch , Germany
| | - Berthold Huppertz
- d Institute of Cell Biology, Histology and Embryology, Medical University of Graz , Graz , Austria
| | - Stefan Verlohren
- a Department of Obstetrics , Charité University Medicine , Berlin , Germany
| |
Collapse
|
38
|
Rätsep MT, Hickman AF, Croy BA. The Elsevier trophoblast research award lecture: Impacts of placental growth factor and preeclampsia on brain development, behaviour, and cognition. Placenta 2016; 48 Suppl 1:S40-S46. [PMID: 26880207 DOI: 10.1016/j.placenta.2016.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 01/19/2016] [Accepted: 02/03/2016] [Indexed: 12/17/2022]
Abstract
Preeclampsia (PE) is a significant gestational disorder affecting 3-5% of all human pregnancies. In many PE pregnancies, maternal plasma is deficient in placental growth factor (PGF), a placentally-produced angiokine. Beyond immediate fetal risks associated with acute termination of the pregnancy, offspring of PE pregnancies (PE-F1) have higher long-term risks for hypertension, stroke, and cognitive impairment compared to F1s from uncomplicated pregnancies. At present, mechanisms that explain PE-F1 gains in postpartum risks are poorly understood. Our laboratory found that mice genetically-deleted for Pgf have altered fetal and adult brain vascular development. This is accompanied by sexually dimorphic alterations in anatomic structure in the adult Pgf-/- brain and impaired cognitive functions. We hypothesize that cerebrovascular and neurological aberrations occur in fetuses exposed to the progressive development of PE and that these brain changes impair cognitive functioning, enhance risk for stroke, elevate severity of stroke, and lead to worse stroke outcomes. These brain and placental outcomes may be linked to down-regulated PGF gene expression in early pre-implantation embryos, prior to gastrulation. This review explores our hypothesis that there are mechanistic links between low PGF detection in maternal plasma prodromal to PE, PE, and altered brain vascular, structural, and functional development amongst PE-F1s. We also include a summary of preliminary outcomes from a pilot study of 7-10 year old children that is the first to report magnetic resonance imaging, magnetic resonance angiography, and functional brain region assessment by eye movement control studies in PE-F1s.
Collapse
Affiliation(s)
- Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| | - Andrew F Hickman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
39
|
Wallenstein MB, Jelliffe-Pawlowski LL, Yang W, Carmichael SL, Stevenson DK, Ryckman KK, Shaw GM. Inflammatory biomarkers and spontaneous preterm birth among obese women. J Matern Fetal Neonatal Med 2016; 29:3317-22. [PMID: 26700828 PMCID: PMC5108178 DOI: 10.3109/14767058.2015.1124083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To identify associations between second-trimester serum inflammatory biomarkers and preterm birth among obese women. METHODS In this nested case-control study, we compared 65 serum inflammatory biomarkers in obese women whose pregnancies resulted in early spontaneous preterm birth (<32 weeks gestation, n = 34) to obese women whose pregnancies resulted in term birth (n = 34). These women were selected from a larger population-based California cohort. Random forest and classification and regression tree techniques were employed to identify biomarkers of importance, and adjusted odds ratios (aORs) and 95% confidence intervals (CI) were estimated using logistic regression. RESULTS Random forest and classification and regression tree techniques found that soluble vascular endothelial growth factor receptor-3 (sVEGFR3), soluble interleukin-2 receptor alpha-chain (sIL-2RA) and soluble tumor necrosis factor receptor-1 (sTNFR1) were related to preterm birth. Using multivariable logistic regression to compare preterm cases and term controls, decreased serum levels of sVEGFR3 and increased serum levels of sIL-2RA and sTNFR1 were associated with increased risk of preterm birth among obese women, aOR = 3.2 (95% CI: 1.0-9.9), aOR = 2.8 (95% CI: 0.9-9.0), and aOR = 4.1 (95% CI: 1.2-14.1), respectively. CONCLUSIONS In this pilot study, we identified three serum biomarkers indicative of inflammation to be associated with spontaneous preterm birth among obese women: sVEGFR3, sIL-2RA and sTNFR1.
Collapse
Affiliation(s)
- Matthew B. Wallenstein
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | - Laura L. Jelliffe-Pawlowski
- Genetic Disease Screening Program, California Department of Public Health, Richmond, CA
- Department of Epidemiology and Biostatistics, University of California San Francisco School of Medicine, San Francisco, CA
| | - Wei Yang
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | - Suzan L. Carmichael
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | - David K. Stevenson
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | | | - Gary M. Shaw
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| |
Collapse
|
40
|
Akarsu S, Bagirzade M, Omeroglu S, Büke B. Placental vascularization and apoptosis in Type-1 and gestational DM. J Matern Fetal Neonatal Med 2016; 30:1045-1050. [DOI: 10.1080/14767058.2016.1199676] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
41
|
Salimi S, Hajizadeh A, Yaghmaei M, Rezaie S, Shahrakypour M, Teimoori B, Parache M, Naghavi A, Mokhtari M. The effects of p21 gene C98A polymorphism on development of uterine leiomyoma in southeast Iranian women. Tumour Biol 2016; 37:12497-12502. [PMID: 27337955 DOI: 10.1007/s13277-016-5078-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 05/15/2016] [Indexed: 12/12/2022] Open
Abstract
Uterine leiomyoma (UL) is a monoclonal tumor which arises from uninhibited proliferation of a single myometrial cell; therefore, the imbalance in cell cycle regulation could be a key event in its development. In the present study, we aimed to assess the association of p21 gene polymorphisms and UL. Genomic DNA was extracted from blood samples of 154 women with UL and 197 age-, BMI-, and ethnically matched controls. p21 C98A (rs1801270) and C70T (rs1059234) polymorphism genotypes were determined by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis. The CA genotype of p21 C98A polymorphism was significantly higher in UL women (28 %) compared to the controls (18 %), and the UL risk was 1.8-fold greater in women with CA genotype compared to CC genotype before and after adjusting for age, BMI, and ethnicity (OR, 1.8 [95 % CI, 1.1 to 3]; P = 0.02). There was no association between the AA genotype of p21 C98A polymorphism and UL. Moreover, the frequency of p21 98A allele was significantly higher in the UL women compared to controls (17 vs. 12 %, p = 0.04). The p21 C70T polymorphism did not correlate with UL before and after adjusting for age, BMI, and ethnicity. There was no difference in haplotype frequency of p21 C70T and C98A polymorphisms between UL patients and the controls. CA genotype of p21 C98A polymorphism may be a risk factor for UL susceptibility; however, p21 C70T polymorphism did not associate with UL.
Collapse
Affiliation(s)
- Saeedeh Salimi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.,Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Azam Hajizadeh
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Minoo Yaghmaei
- Department of Obstetrics and Gynecology, School of Medicine, Shahid Beheshty University of Medical Sciences, Tehran, Iran.
| | - Sodabeh Rezaie
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahnaz Shahrakypour
- Department of Biostatistics and Epidemiology, School of Public Health, Zahedan University of Medical Sciences, Zahedan, Iran.,Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Batool Teimoori
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahboube Parache
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Anoosh Naghavi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mojgan Mokhtari
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
42
|
Vascular endothelial growth factor and its receptors in the placenta of women with type 1 diabetes mellitus. Bull Exp Biol Med 2016; 152:367-70. [PMID: 22803088 DOI: 10.1007/s10517-012-1530-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We performed a morphological study of placentas from women with type 1 diabetes mellitus receiving insulin therapy (insulin pump). Expression of vascular endothelial growth factor (VEGF) and its receptors (VEGFR-1, VEGFR-2, VEGFR-3) was demonstrated by immunohistochemical methods. Processes of branched angiogenesis predominated in the placentas from women with type 1 diabetes mellitus. Immunohistochemical study revealed more intensive reaction of VEGF and its receptors in syncytiotrophoblast and capillary endothelium of terminal villi.
Collapse
|
43
|
Lee H, Kwon JY, Lee S, Kim SJ, Shin JC, Park IY. Elevated placenta growth factor levels in the early second-trimester amniotic fluid are associated with preterm delivery. J Matern Fetal Neonatal Med 2016; 29:3374-8. [PMID: 26634962 DOI: 10.3109/14767058.2015.1127345] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVE The purpose of this study was to determine how angiogenesis-related factors correlate with preterm delivery. METHODS A cohort of 382 pregnant women undergoing early second-trimester genetic amniocentesis was enrolled and followed-up until delivery, and the amniotic fluid was collected and stored as a nested case-control study. Cases with preterm delivery (n = 31) were compared with matched controls with term delivery (n = 62). The amniotic fluid concentrations of placenta growth factor (PlGF), angiogenins, angiopoietin-2, soluble fms-like tyrosine kinase and soluble endoglin were determined using enzyme-linked immunosorbent assays. RESULTS Women who delivered preterm had a higher amniotic PlGF concentration compared with the control group (median 12.6 pg/ml versus 6.1 pg/ml; p=0.027). Other angiogenesis-related factors did not show any differences between case and control groups. The odds ratio for preterm delivery based on amniotic fluid PlGF was 1.031 (95% confidence interval: 1.002-1.061; p=0.035). Additionally, when the cases were subdivided into early preterm, late preterm and term groups, PlGF values between the early preterm and term delivery groups were significantly different (median 16.6 pg/ml versus 6.1 pg/ml; Bonferroni-adjusted p=0.018). CONCLUSION Amniotic fluid PlGF levels in the early second trimester of pregnancy are associated with preterm delivery.
Collapse
Affiliation(s)
- Hambak Lee
- a Department of Obstetrics and Gynecology, College of Medicine , the Catholic University of Korea , Seoul , Korea and
| | - Ji Young Kwon
- a Department of Obstetrics and Gynecology, College of Medicine , the Catholic University of Korea , Seoul , Korea and
| | - Seungok Lee
- b Department of Laboratory Medicine , College of Medicine, the Catholic University of Korea , Seoul , Korea
| | - Sa Jin Kim
- a Department of Obstetrics and Gynecology, College of Medicine , the Catholic University of Korea , Seoul , Korea and
| | - Jong Chul Shin
- a Department of Obstetrics and Gynecology, College of Medicine , the Catholic University of Korea , Seoul , Korea and
| | - In Yang Park
- a Department of Obstetrics and Gynecology, College of Medicine , the Catholic University of Korea , Seoul , Korea and
| |
Collapse
|
44
|
Kim J, Lee J, Kim SH, Jun JH. Coculture of Preimplantation Embryos With Outgrowth Embryos Improves Embryonic Developmental Competence in Mice. Reprod Sci 2015; 23:913-23. [DOI: 10.1177/1933719115623641] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jihyun Kim
- Department of Senior Healthcare, BK21 Plus Program, Graduated School, Eulji University, Daejeon, Korea
| | - Jaewang Lee
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Korea
- Seoul National University College of Medicine, Seoul, Korea
| | - Seok Hyun Kim
- Seoul National University College of Medicine, Seoul, Korea
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, Korea
| | - Jin Hyun Jun
- Department of Senior Healthcare, BK21 Plus Program, Graduated School, Eulji University, Daejeon, Korea
- Department of Biomedical Laboratory Science, Graduate School of Health Science, Eulji University, Daejeon, Korea
- Eulji Medi-Bio Research Institute (EMBRI), Eulji University, Daejeon, Korea
| |
Collapse
|
45
|
Yaghmaei M, Salimi S, Namazi L, Farajian-Mashhadi F. Association of XRCC1 Arg399GIn and Tp53 Arg72Pro polymorphisms and increased risk of uterine leiomyoma - A case-control study. Genet Mol Biol 2015; 38:444-9. [PMID: 26692154 PMCID: PMC4763320 DOI: 10.1590/s1415-475738420140359] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/12/2015] [Indexed: 02/26/2023] Open
Abstract
The aim of present study was to investigate the role of the X-ray repair cross-complementing protein1 (XRCC1) and Tumor protein p53 (Tp53) polymorphisms in Uterine Leiomyoma (UL) susceptibility in southeastern Iran. This case control study was performed on 139 women with UL and 149 age, BMI and ethnicity matched healthy women. All women were genotyped for the XRCC1 Arg399Gln, XRCC1 Arg194Trp and Tp53 Arg72Pro polymorphisms. The frequency of Tp53 72 Pro/Pro genotype was significantly higher in UL women compared to controls. The risk of UL was 1.5 fold higher in women with the Pro/Pro genotype (OR, 1.5 [95% CI, 1.1 to 2.1], p = 0.012). Moreover, the frequency of the Pro allele was significantly higher in the UL women. Although the frequency of XRCC1 Arg399Gln genotypes did not significantly differ between UL and control groups before adjusting for age, there was an association between the XRCC1 Arg/Gln genotype and UL after adjusting for age (OR, 1.8 [95% CI, 1.1 to 3]). No association was observed between the XRCC1 Arg194Trp polymorphism and UL. The Pro/Pro genotype of Tp53 Arg72Pro polymorphism was associated with UL susceptibility. In addition, the XRCC1 Arg/Gln genotype was associated with increased risk of UL after adjusting for age.
Collapse
Affiliation(s)
- Minoo Yaghmaei
- Department of Obstetrics and Gynecology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeedeh Salimi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Lida Namazi
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | | |
Collapse
|
46
|
Salimi S, Yaghmaei M, Tabatabaei E, Mokhtari M, Naghavi A. Vascular endothelial growth factor (VEGF)-634G/C polymorphism was associated with severe pre-eclampsia and lower serum VEGF level. J Obstet Gynaecol Res 2015; 41:1877-83. [PMID: 26486233 DOI: 10.1111/jog.12825] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 06/29/2015] [Accepted: 07/05/2015] [Indexed: 02/02/2023]
Abstract
AIM Vascular endothelial growth factor (VEGF) is an angiogenic factor whose production is increased in pre-eclampsia (PE). Therefore, the present study was conducted aiming at assessing the possible association of VEGF polymorphisms with PE susceptibility in the southeast of Iran. MATERIAL AND METHODS Overall, 192 PE women and 186 unrelated age-matched normotensive pregnant women were genotyped for the VEGF-2578C/A (rs699947), -1154G/A (rs1570360), and -634G/C (rs2010963) polymorphisms using the polymerase chain reaction-restriction fragment length polymorphism method. Serum VEGF levels were determined by the enzyme-linked immunosorbent assay method. RESULTS There was no significant difference in VEGF-2578C/A, -1154G/A and -634G/C polymorphisms between PE women and controls. However, the frequency of VEGF-634GC and CC genotypes was significantly higher in women with severe PE compared to mild PE and controls. In addition, serum VEGF levels were significantly lower in PE women. The VEGF-634CC genotype was associated with lower serum VEGF levels compared to the VEGF-634GG genotype. Moreover, serum VEGF levels were significantly lower in individuals with the VEGF-634CC genotype compared to VEGF-634GC genotype only in the control group. The mean serum VEGF levels did not differ significantly between genotypes of VEGF-2587C/A and -1154G/A polymorphisms. CONCLUSION Our findings suggest that the association of VEGF-634G/C polymorphisms with severe PE and the VEGF-634CC genotype was correlated with lower serum VEGF levels.
Collapse
Affiliation(s)
- Saeedeh Salimi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.,Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Minoo Yaghmaei
- Department of Obstetrics and Gynecology, School of Medicine, Shahid Beheshty University of Medical Sciences, Tehran, Iran
| | - Ehsan Tabatabaei
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mojgan Mokhtari
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.,Pregnancy Health Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Anoosh Naghavi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.,Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| |
Collapse
|
47
|
Li C, Raikwar NS, Santillan MK, Santillan DA, Thomas CP. Aspirin inhibits expression of sFLT1 from human cytotrophoblasts induced by hypoxia, via cyclo-oxygenase 1. Placenta 2015; 36:446-53. [PMID: 25638730 DOI: 10.1016/j.placenta.2015.01.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 12/29/2014] [Accepted: 01/08/2015] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Elevated circulating soluble FLT1 (sFLT1) levels seen in preeclampsia may play a role in its development. Aspirin is recommended for prevention of preeclampsia. We hypothesized that aspirin may inhibit the production of sFlt1. METHODS Placentas from women with and without preeclampsia were collected. Primary cytotrophoblasts (CTBs) were cultured from normal placentas and treated with aspirin, sc-560, a COX1 inhibitor or celecoxib, a COX2 inhibitor. The expression of sFLT1, FLT1, COX1 and COX2 was studied. The effect of aspirin on sFlt1 expression was also studied in HEK293 cells and in HTR-8/SVNeo cells. RESULTS The expression of sFLT1 was increased in preeclamptic placentas compared to control placentas and the expression and release of sFLT1 increased in CTBs exposed to 2% O2 compared to controls. Aspirin at 3 and 12 mM concentration reduced the expression and release of sFLT1 in CTBs. Aspirin also inhibited sFlt1 expression from HTR-8/SVNeo and HEK293 cells. Sc-560, but not celecoxib, reduced sFLT1 expression and release from CTBs. Aspirin and sc-560 also reduced hypoxia-induced FLT1 mRNA expression and inhibited COX1 mRNA in CTBs. DISCUSSION This study confirms that sFLT1 expression is increased in preeclamptic placentas and in CTBs exposed to hypoxia. Aspirin inhibits the production sFLT1 in CTBs and in HTR-8/SVNeo. Sc-560 recapitulated the effects of aspirin on sFLT1 expression and release in CTBs suggesting that the aspirin effect may be mediated via inhibition of COX1. The study increases our understanding of the mechanisms regulating sFlt1 expression and provides a plausible explanation for the effect of aspirin to prevent preeclampsia.
Collapse
Affiliation(s)
- C Li
- Department of Obstetrics and Gynecology, Iowa City, IA, USA
| | - N S Raikwar
- Division of Nephrology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - M K Santillan
- Department of Obstetrics and Gynecology, Iowa City, IA, USA
| | - D A Santillan
- Department of Obstetrics and Gynecology, Iowa City, IA, USA
| | - C P Thomas
- Department of Obstetrics and Gynecology, Iowa City, IA, USA; Division of Nephrology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA; Department of Veterans Affairs Medical Center, Iowa City, IA, USA.
| |
Collapse
|
48
|
Li Y, Zhao YJ, Zou QY, Zhang K, Wu YM, Zhou C, Wang K, Zheng J. Preeclampsia does not alter vascular growth and expression of CD31 and vascular endothelial cadherin in human placentas. J Histochem Cytochem 2014; 63:22-31. [PMID: 25362142 DOI: 10.1369/0022155414558063] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Preeclampsia is characterized by maternal endothelial dysfunction (e.g., increased maternal vascular permeability caused by the disassembly of endothelial junction proteins). However, it is unclear if preeclampsia is associated with impaired vascular growth and expression of endothelial junction proteins in human placentas. Herein, we examined vascular growth in placentas from women with normal term (NT) and preeclamptic (PE) pregnancies using two endothelial junction proteins as endothelial markers: CD31 and vascular endothelial-cadherin (VE-Cad). We also compared protein and mRNA expression of CD31 and VE-Cad between NT and PE placentas, and determined the alternatively spliced expression of CD31 using PCR. We found that CD31 and VE-Cad were immunolocalized predominantly in villous endothelial cells. However, capillary number density (total capillary number per unit villous area) and capillary area density (total capillary lumen area per unit villous area) as well as CD31 and VE-Cad protein and mRNA levels were similar between NT and PE placentas. PCR in combination with sequence analysis revealed a single, full-length CD31, suggesting that there are no alternatively spliced isoform of CD31 expressed in placentas. These data indicate that preeclampsia does not significantly affect vascular growth or the expression of endothelial junction proteins in human placentas.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ)
| | - Ying-Jie Zhao
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ),Department of Rheumatology, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China (YJZ)
| | - Qing-Yun Zou
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ)
| | - Kevin Zhang
- Department of Biological Sciences, Dartmouth College, Hanover, NH (KZ)
| | - Yan-Ming Wu
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China (YMW, KW)
| | - Chi Zhou
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ)
| | - Kai Wang
- Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China (YMW, KW)
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, Wisconsin (YL, YJZ, QYZ, CZ, JZ),Department of Cardiovascular Medicine, Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, P.R. China (JZ)
| |
Collapse
|
49
|
Liao TL, Chen SC, Tzeng CR, Kao SH. TCDD induces the hypoxia-inducible factor (HIF)-1α regulatory pathway in human trophoblastic JAR cells. Int J Mol Sci 2014; 15:17733-50. [PMID: 25272228 PMCID: PMC4227186 DOI: 10.3390/ijms151017733] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 09/19/2014] [Accepted: 09/22/2014] [Indexed: 02/06/2023] Open
Abstract
The exposure to dioxin can compromise pregnancy outcomes and increase the risk of preterm births. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) has been demonstrated to induce placental hypoxia at the end of pregnancy in a rat model, and hypoxia has been suggested to be the cause of abnormal trophoblast differentiation and placental insufficiency syndromes. In this study, we demonstrate that the non-hypoxic stimulation of human trophoblastic cells by TCDD strongly increased hypoxia inducible factor-1 alpha (HIF-1α) stabilization. TCDD exposure induced the generation of reactive oxygen species (ROS) and nitric oxide. TCDD-induced HIF-1α stabilization and Akt phosphorylation was inhibited by pretreatment with wortmannin (a phosphatidylinositol 3-kinase (PI3K) inhibitor) or N-acetylcysteine (a ROS scavenger). The augmented HIF-1α stabilization by TCDD occurred via the ROS-dependent activation of the PI3K/Akt pathway. Additionally, a significant increase in invasion and metallomatrix protease-9 activity was found in TCDD-treated cells. The gene expression of vascular endothelial growth factor and placental growth factor was induced upon TCDD stimulation, whereas the protein levels of peroxisome proliferator-activated receptor γ (PPARγ), PPARγ coactivator-1α, mitochondrial transcription factor, and uncoupling protein 2 were decreased. Our results indicate that an activated HIF-1α pathway, elicited oxidative stress, and induced metabolic stress contribute to TCDD-induced trophoblastic toxicity. These findings may provide molecular insight into the TCDD-induced impairment of trophoblast function and placental development.
Collapse
Affiliation(s)
- Tien-Ling Liao
- Graduate Institute of Medical Science, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Su-Chee Chen
- Department of Obstetrics and Gynecology, Cathay General Hospital, Taipei 110, Taiwan.
| | - Chii-Reuy Tzeng
- Center for Reproductive Medicine & Sciences Taipei Medical University Hospital, Taipei 110, Taiwan.
| | - Shu-Huei Kao
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
50
|
Glucose, insulin, and oxygen interplay in placental hypervascularisation in diabetes mellitus. BIOMED RESEARCH INTERNATIONAL 2014; 2014:145846. [PMID: 25258707 PMCID: PMC4167234 DOI: 10.1155/2014/145846] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/06/2014] [Indexed: 02/07/2023]
Abstract
The placental vasculature rapidly expands during the course of pregnancy in order to sustain the growing needs of the fetus. Angiogenesis and vascular growth are stimulated and regulated by a variety of growth factors expressed in the placenta or present in the fetal circulation. Like in tumors, hypoxia is a major regulator of angiogenesis because of its ability to stimulate expression of various proangiogenic factors. Chronic fetal hypoxia is often found in pregnancies complicated by maternal diabetes as a result of fetal hyperglycaemia and hyperinsulinemia. Both are associated with altered levels of hormones, growth factors, and proinflammatory cytokines, which may act in a proangiogenic manner and, hence, affect placental angiogenesis and vascular development. Indeed, the placenta in diabetes is characterized by hypervascularisation, demonstrating high placental plasticity in response to diabetic metabolic derangements. This review describes the major regulators of placental angiogenesis and how the diabetic environment in utero alters their expression. In the light of hypervascularized diabetic placenta, the focus was placed on proangiogenic factors.
Collapse
|