1
|
Wang H, Fan N, Cui X, Xie R, Tang Y, Thomas AM, Li S, Zhang JV, Liu S, Qin H. BMP5 promotes trophoblast functions upon N-glycosylation via the BMP5-SMAD1/5 signaling pathway in preeclampsia. Placenta 2024; 158:240-252. [PMID: 39520832 DOI: 10.1016/j.placenta.2024.11.002] [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: 07/14/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Preeclampsia (PE) is one of the most common pregnancy-related complications worldwide and currently lacks an effective treatment. While trophoblast cell dysfunction has been identified as the fundamental cause of PE, the underlying mechanisms remain unclear. Bone morphogenetic protein 5 (BMP5) is a secreted glycoprotein highly expressed in the placenta that is involved in cell proliferation, migration, and invasion. However, the role and mechanism of BMP5 glycosylation of trophoblast cell function remain unclear. METHODS The expression of BMP5 and N-glycosylation in preeclamptic placental tissues was investigated. We predicted and validated the N-glycosylation sites of BMP5. Additionally, we evaluated the effect of BMP5 N-glycosylation on the proliferation, migration, invasion, and angiogenesis of human immortalized trophoblastic HTR-8/SVneo cells. Furthermore, the role of N-glycosylated BMP5 in activating the BMP5-SMAD1/5 signaling pathway and regulating trophoblastic cell functions was explored. RESULTS Our study reveals that PHA-E + L (recognizing branching N-glycans) reactive N-glycans and BMP5 expression levels are lower in preeclamptic villous tissues compared to normal placental tissues. Additionally, we demonstrated that BMP5 is an N-glycosylation-modified protein. Furthermore, N-glycosylated BMP5 promoted the functional trophoblastic cells (HTR-8/SVneo). We also revealed that N-glycosylation of BMP5 regulates multiple cell functions through the BMP5-SMAD1/5 signaling pathway. CONCLUSION N-glycosylated BMP5 promotes trophoblast cell proliferation, migration, invasion, and angiogenesis. This study provides mechanistic insight as to how N-glycosylation of BMP5 in trophoblast cells can contribute to the pathogenesis of preeclampsia and provides a new basis for its diagnosis and treatment.
Collapse
Affiliation(s)
- Hao Wang
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China; Sino-European Center of Biomedicine and Health, Shenzhen, China
| | - Ningning Fan
- Department of Laboratory Medicine, The First Affiliated Hospital of Northwest University, Xi'an No.1 Hospital, Xi'an, China; Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xinyuan Cui
- The University of Hong Kong Shenzhen Key Laboratory of Fertility Regulation, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ru Xie
- Department of Pathology, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Ying Tang
- Department of Pathology, The Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Aline M Thomas
- The Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jian V Zhang
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, China; Sino-European Center of Biomedicine and Health, Shenzhen, China
| | - Shuai Liu
- Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, College of Basic Medical Science, Dalian Medical University, Dalian, China.
| | - Huamin Qin
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
2
|
Bukowska P, Bralewska M, Pietrucha T, Sakowicz A. Nutraceuticals as Modulators of Molecular Placental Pathways: Their Potential to Prevent and Support the Treatment of Preeclampsia. Int J Mol Sci 2024; 25:12167. [PMID: 39596234 PMCID: PMC11594370 DOI: 10.3390/ijms252212167] [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: 10/31/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Preeclampsia (PE) is a serious condition characterized by new-onset hypertension and proteinuria or organ dysfunction after the 20th week of gestation, making it a leading cause of maternal and fetal mortality worldwide. Despite extensive research, significant gaps remain in understanding the mechanisms underlying PE, contributing to the ineffectiveness of current prevention and treatment strategies. Consequently, premature cesarean sections often become the primary intervention to safeguard maternal and fetal health. Emerging evidence indicates that placental insufficiency, driven by molecular disturbances, plays a central role in the development of PE. Additionally, the maternal microbiome may be implicated in the pathomechanism of preeclampsia by secreting metabolites that influence maternal inflammation and oxidative stress, thereby affecting placental health. Given the limitations of pharmaceuticals during pregnancy due to potential risks to fetal development and concerns about teratogenic effects, nutraceuticals may provide safer alternatives. Nutraceuticals are food products or dietary supplements that offer health benefits beyond basic nutrition, including plant extracts or probiotics. Their historical use in traditional medicine has provided valuable insights into their safety and efficacy, including for pregnant women. This review will examine how the adoption of nutraceuticals can enhance dysregulated placental pathways, potentially offering benefits in the prevention and treatment of preeclampsia.
Collapse
Affiliation(s)
| | | | | | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
3
|
Tan S, Tan S, Tokgün O, Çetin H, Tokgün E, Özdamar S. Investigation of diabetes-related molecular changes in embryo-endometrium crosstalk. Gene 2024; 922:148557. [PMID: 38740354 DOI: 10.1016/j.gene.2024.148557] [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: 11/16/2023] [Revised: 03/18/2024] [Accepted: 05/10/2024] [Indexed: 05/16/2024]
Abstract
The primary aim of this study was to explore the impact of diabetes on matrix metalloproteases and tissue inhibitors, crucial factors for successful implantation, and to elucidate the molecular mechanisms that undergo changes in the endometrium and the embryo during diabetic pregnancies. In this investigation, we established a streptozotocin-induced diabetic pregnant rat model. Microarray analysis followed by RT-PCR was utilized to identify gene regions exhibiting expression alterations. Subsequently, we assessed the effects of MMPs and tissue inhibitors using ELISA and immunohistochemistry techniques, in addition to analyzing changes at the genetic level. Diabetes led to the upregulation of MMP3, MMP9, and MMP20 on the 6.5th day of pregnancy, while causing the downregulation of MMP3, MMP9, and MMP11 on the 8.5th day of pregnancy. TIMP1 expression was downregulated on the 8.5th day compared to the control group. No statistically significant differences were observed between the groups regarding other TIMP expressions. KEGG pathway analysis revealed that diabetes induced alterations in the expression of genes associated with certain microRNAs, as well as signaling pathways such as cAMP, calcium, BMP, p53, MAPK, PI3K-Akt, Jak-STAT, Hippo, Wnt, and TNF. Additionally, gene ontology analysis unveiled changes in membrane structures, extracellular matrix, signaling pathways, ion binding, protein binding, cell adhesion molecule binding, and receptor-ligand activity. This study serves as a valuable guide for investigating the mechanisms responsible for complications in diabetic pregnancies. By revealing the early-stage effects of diabetes, it offers insight into the development of new diagnostic and treatment approaches, ultimately contributing to improved patient care.
Collapse
Affiliation(s)
- Semih Tan
- Ordu University, Faculty of Medicine, Department of Histology and Embryology, Turkiye.
| | - Seçil Tan
- Pamukkale University, Faculty of Medicine, Department of Medical Genetic, Turkiye
| | - Onur Tokgün
- Pamukkale University, Faculty of Medicine, Department of Medical Genetic, Turkiye
| | - Hülya Çetin
- Pamukkale University, Faculty of Medicine, Department of Histology and Embryology, Turkiye
| | - Elvan Tokgün
- Pamukkale University, Faculty of Medicine, Department of Medical Genetic, Turkiye
| | - Saim Özdamar
- Pamukkale University, Faculty of Medicine, Department of Histology and Embryology, Turkiye
| |
Collapse
|
4
|
Sahin E, Eraslan Sahin M, Kirlangic MM, Kutuk S, Daglıtuncezdi Cam S, Can Ozdemir H, Genc E. Effects of different PCOS phenotypes on placental three-dimensional power Doppler indices and placental volume during the first trimester. Placenta 2024; 154:176-183. [PMID: 39018610 DOI: 10.1016/j.placenta.2024.07.002] [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: 02/09/2024] [Revised: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024]
Abstract
INTRODUCTION The aim of the present study was to evaluate the effects of different polycystic ovary syndrome (PCOS) phenotypes using first-trimester placental three-dimensional power Doppler indices and placental volume. METHODS In this prospective case-control study, 170 pregnant women who met the inclusion criteria were divided into five groups according to PCOS phenotype: non-PCOS control (n = 34), PCOS phenotype A (n = 34), PCOS phenotype B (n = 34), PCOS phenotype C (n = 34), and PCOS phenotype D (n = 34). The primary outcomes determined in the present study were the differences in placental volume and placental flow index (FI), vascularization flow index (VFI), vascularization index (VI), and uterine artery pulsatility index (PI) betweenthe PCOS groups and control group. RESULTS The mean placental volume and VI were significantly decreased in the phenotype A, B, and C groups compared to the control group and PCOS phenotype D group. The mean FI and VFI were significantly decreased in the phenotype A and B groups compared to the control group and PCOS phenotype C and D groups. The mean testosterone, dehydroepiandrostenedione, sex-hormone binding globulin, free androgen index, and insulin resistance levels were significantly increased in the phenotype A, B, and C groups compared to the control group and PCOS phenotype D group. DISCUSION The results indicated that placental volume and placental vascular Doppler indices in the first trimester were more adversely affected in the PCOS A and B phenotypes than other PCOS phenotypes.
Collapse
Affiliation(s)
- Erdem Sahin
- Department of Obstetrics and Gynecology, Kayseri City Hospital, Kayseri, Turkey.
| | | | - Mehmet Mete Kirlangic
- Department of Obstetrics and Gynecology, Istanbul Dr. Lutfu Kirdar City Hospital, Istanbul, Turkey
| | - Serhan Kutuk
- Department of Obstetrics and Gynecology, Kayseri Develi State Hospital, Kayseri, Turkey
| | | | - Havva Can Ozdemir
- Department of Obstetrics and Gynecology, Kayseri City Hospital, Kayseri, Turkey
| | - Emre Genc
- Department of Obstetrics and Gynecology, Kayseri City Hospital, Kayseri, Turkey
| |
Collapse
|
5
|
Ramdin S, Naicker T, Baijnath S, Govender N. Is renal dysfunction amplified in an arginine vasopressin induced rat model of preeclampsia? Reprod Biol 2024; 24:100910. [PMID: 38851025 DOI: 10.1016/j.repbio.2024.100910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 04/27/2024] [Accepted: 05/25/2024] [Indexed: 06/10/2024]
Abstract
Renal dysfunction is important in preeclampsia (PE) pathophysiology and has not been fully explored in the arginine vasopressin (AVP) rat model of PE. This study aimed to determine kidney toxicity associated with this model. Female Sprague Dawley rats (n = 24) were subcutaneously infused with AVP or saline for 18 days. Urine samples (GD8, 14 and 18) were used to determine the levels of albumin, VEGF-A, clusterin, NGAL/Lipocalin-2, KIM-1, cystatin C, TIMP-1, β2M and OPN via Multiplex ELISAs. Albumin, and NGAL/lipocalin-2 were significantly elevated in the PAVP vs PS group on GD14 and GD18 (p < 0.001) respectively. VEGF-A significantly decreased in the pregnant vs non-pregnant groups on GD14 and 18 (p < 0.001). Clusterin (p < 0.001) and OPN (p < 0.05) were significantly higher in the PAVP vs PS group on GD18. Cystatin C and KIM-1 are significantly upregulated in the PAVP vs PS groups throughout gestation (p < 0.05). β2M is significantly elevated in the PAVP vs PS group on GD14 and 18 (p < 0.05). AVP elevated the urinary levels of the kidney injury biomarkers and replicated the renal dysfunction associated with PE development. Our findings confirm the potential applications of this model in studying the mechanisms underlying renal damage in PE.
Collapse
Affiliation(s)
- Sapna Ramdin
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institute, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sooraj Baijnath
- Integrated Molecular Physiology Research Initiative, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nalini Govender
- Department of Basic Medical Sciences, Faculty of Health Sciences, Durban University of Technology, Durban, South Africa.
| |
Collapse
|
6
|
Paiva RVN, Mondes PHDL, Brandão BDJ, Sant’Anna JN, Freire dos Santos ME, Fighera YM, Santos LC, Markus RP, Fernandes PACM, Silva JF, Tamura EK. Effects of acute hypothyroidism on plasma melatonin and Aanat and Asmt expression in the pineal gland and gonads of rats. Front Endocrinol (Lausanne) 2024; 15:1322867. [PMID: 39149125 PMCID: PMC11324505 DOI: 10.3389/fendo.2024.1322867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/01/2024] [Indexed: 08/17/2024] Open
Abstract
Introduction The reproductive system is tightly regulated by environmental and physiological signals. Melatonin, known as the hormone of darkness, plays a crucial role in regulating both the circadian and reproductive systems in mammals. Hypothyroidism is a key endocrine disorder that harms the reproductive system. Despite many studies on melatonin's effects on the reproductive system, there is conflicting information regarding melatonin synthesis modulation in hypothyroidism. The objective of this study was to investigate the modulation of plasma melatonin levels and gene expression of Aanat and Asmt in the pineal gland and gonads of rats with hypothyroidism at different times of the day. Methods Female and male Wistar rats were divided into control and hypothyroid groups. Hypothyroidism was induced using propylthiouracil (PTU) for 15 days, rats were euthanized six hours after lights on (ZT6), before lights off (ZT11.5), and six hours after lights off (ZT18). Free thyroxine (FT4) and melatonin were quantified in plasma, and gene expressions of melatonin synthesizing enzymes (Aanat and Asmt) were measured in pineal and sexual organs (testis and ovary). Also, morphological analysis was performed in sexual organs. Results The results reveal some disparities between the sexes. Hypothyroidism reduced antral and primary follicles in the ovary, and reduced the weight of testis, epididymis, and prostate. In relation to gene expression, we observed a reduction in Aanat expression in the pineal gland during the light phase (ZT6), and in males, this reduction occurred during the dark phase (ZT18). Regarding Asmt expression, there was a decrease in females also during the dark phase (ZT18). In the gonads, there was an increase in expression in both sexes at ZT11.5. Additionally, it was interesting to observe the association between FT4 levels and Asmt expression in the gonads. Conclusions This study showed that acute hypothyroidism can affect components of the melatonergic system in gonads, particularly gene expression of melatonin synthesis enzymes (Aanat and Asmt) contributing to changes in reproduction organs during disease progression. These findings enhance our understanding of melatonin synthesis in the reproductive system during hypothyroidism, showing distinct responses in male and female rats, and suggest that hypothyroidism affects the circadian rhythmicity of melatonin synthesis in a sex-dependent manner.
Collapse
Affiliation(s)
- Rafaella Valete Nunes Paiva
- Chronobiology Research Group, Department of Health Sciences, State University of Santa Cruz, Ilhéus, Brazil
- Reproduction and Endocrinology Research Center, Department of Biological Sciences, State University of Santa Cruz, Ilhéus, Brazil
| | | | - Beatriz de Jesus Brandão
- Chronobiology Research Group, Department of Health Sciences, State University of Santa Cruz, Ilhéus, Brazil
| | - Julia Nascimento Sant’Anna
- Chronobiology Research Group, Department of Health Sciences, State University of Santa Cruz, Ilhéus, Brazil
| | | | - Yasmin Muniz Fighera
- Chronobiology Research Group, Department of Health Sciences, State University of Santa Cruz, Ilhéus, Brazil
| | - Luciano Cardoso Santos
- Reproduction and Endocrinology Research Center, Department of Biological Sciences, State University of Santa Cruz, Ilhéus, Brazil
| | - Regina P. Markus
- Chronopharmacology Laboratory, Department of Physiology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Juneo Freitas Silva
- Reproduction and Endocrinology Research Center, Department of Biological Sciences, State University of Santa Cruz, Ilhéus, Brazil
| | - Eduardo Koji Tamura
- Chronobiology Research Group, Department of Health Sciences, State University of Santa Cruz, Ilhéus, Brazil
| |
Collapse
|
7
|
Li H, Miao D, Hu H, Xue P, Zhou K, Mao Z. Titanium Dioxide Nanoparticles Induce Maternal Preeclampsia-like Syndrome and Adverse Birth Outcomes via Disrupting Placental Function in SD Rats. TOXICS 2024; 12:367. [PMID: 38787146 PMCID: PMC11125676 DOI: 10.3390/toxics12050367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024]
Abstract
The escalating utilization of titanium dioxide nanoparticles (TiO2 NPs) in everyday products has sparked concerns regarding their potential hazards to pregnant females and their offspring. To address these concerns and shed light on their undetermined adverse effects and mechanisms, we established a pregnant rat model to investigate the impacts of TiO2 NPs on both maternal and offspring health and to explore the underlying mechanisms of those impacts. Pregnant rats were orally administered TiO2 NPs at a dose of 5 mg/kg body weight per day from GD5 to GD18 during pregnancy. Maternal body weight, organ weight, and birth outcomes were monitored and recorded. Maternal pathological changes were examined by HE staining and TEM observation. Maternal blood pressure was assessed using a non-invasive blood analyzer, and the urinary protein level was determined using spot urine samples. Our findings revealed that TiO2 NPs triggered various pathological alterations in maternal liver, kidney, and spleen, and induced maternal preeclampsia-like syndrome, as well as leading to growth restriction in the offspring. Further examination unveiled that TiO2 NPs hindered trophoblastic cell invasion into the endometrium via the promotion of autophagy. Consistent hypertension and proteinuria resulted from the destroyed the kidney GBM. In total, an exposure to TiO2 NPs during pregnancy might increase the risk of human preeclampsia through increased maternal arterial pressure and urinary albumin levels, as well as causing fetal growth restriction in the offspring.
Collapse
Affiliation(s)
- Haixin Li
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China; (H.L.); (H.H.); (P.X.)
| | - Dandan Miao
- Huai’an Center for Disease Control and Prevention, Huai’an 223001, China;
| | - Haiting Hu
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China; (H.L.); (H.H.); (P.X.)
| | - Pingping Xue
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China; (H.L.); (H.H.); (P.X.)
| | - Kun Zhou
- State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing 211100, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211100, China
- Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Zhilei Mao
- Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou 213003, China; (H.L.); (H.H.); (P.X.)
- State Key Laboratory of Reproductive Medicine, Center for Global Health, Nanjing Medical University, Nanjing 211100, China
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211100, China
| |
Collapse
|
8
|
Chettiar V, Patel A, Chettiar SS, Jhala DD. Meta-analysis of endometrial transcriptome data reveals novel molecular targets for recurrent implantation failure. J Assist Reprod Genet 2024; 41:1417-1431. [PMID: 38456991 PMCID: PMC11143096 DOI: 10.1007/s10815-024-03077-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 02/27/2024] [Indexed: 03/09/2024] Open
Abstract
PURPOSE Gene expression analysis of the endometrium has been shown to be a useful approach for identifying the molecular signatures and pathways involved in recurrent implantation failure (RIF). Nevertheless, individual studies have limitations in terms of study design, methodology and analysis to detect minor changes in expression levels or identify novel gene signatures associated with RIF. METHOD To overcome this, we conducted an in silico meta-analysis of nine studies, the systematic collection and integration of gene expression data, utilizing rigorous selection criteria and statistical techniques to ensure the robustness of our findings. RESULTS Our meta-analysis successfully unveiled a meta-signature of 49 genes closely associated with RIF. Of these genes, 38 were upregulated and 11 downregulated in RIF patients' endometrium and believed to participate in key processes like cell differentiation, communication, and adhesion. GADD45A, IGF2, and LIF, known for their roles in implantation, were identified, along with lesser-studied genes like OPRK1, PSIP1, SMCHD1, and SOD2 related to female infertility. Many of these genes are involved in MAPK and PI3K-Akt pathways, indicating their role in inflammation. We also investigated to look for key miRNAs regulating these 49 dysregulated mRNAs as potential diagnostic biomarkers. Along with this, we went to associate protein-protein interactions of 49 genes, and we could recognize one cluster consisting of 11 genes (consisted of 22 nodes and 11 edges) with the highest score (p = 0.001). Finally, we validated some of the genes by qRT-PCR in our samples. CONCLUSION In summary, the meta-signature genes hold promise for improving RIF patient identification and facilitating the development of personalized treatment strategies, illuminating the multifaceted nature of this complex condition.
Collapse
Affiliation(s)
- Venkatlaxmi Chettiar
- Department of Life Sciences, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India
| | - Alpesh Patel
- GeneXplore Diagnostics and Research Centre PVT. LTD., Ahmedabad, Gujarat, India
| | | | - Devendrasinh D Jhala
- Department of Zoology, School of Sciences, Gujarat University, Ahmedabad, Gujarat, India.
| |
Collapse
|
9
|
Wang ZG, Yang FL, Liu CY, Wang F, Xiong Y, Zhang Q, Chen MN, Lai H. Predicting intraoperative hemorrhage during curettage treatment of cesarean scar pregnancy using free-breathing GRASP DCE-MRI. BMC Pregnancy Childbirth 2024; 24:22. [PMID: 38172701 PMCID: PMC10763255 DOI: 10.1186/s12884-023-06188-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
OBJECTIVE To explore the feasibility of the golden-angle radial sparse parallel (GRASP) dynamic magnetic resonance imaging (MRI) technique in predicting the intraoperative bleeding risk of scar pregnancy. METHODS A total of 49 patients with cesarean scar pregnancy (CSP) who underwent curettage and GRASP-MRI imaging were retrospectively selected between January 2021 and July 2022. The pharmacokinetic parameters, including Wash-in, Wash-out, time to peck (TTP), initial area under the curve (iAUC), the transfer rate constant (Ktrans), constant flow rate (Kep), and volume of extracellular space (Ve), were calculated. The amount of intraoperative bleeding was recorded by a gynecologist who performed surgery, after which patients were divided into non-hemorrhage (blood loss ≤ 200 mL) and hemorrhage (blood loss > 200 mL) groups. The measured pharmacokinetic parameters were statistically compared using the t-test or Mann-Whitney U test with a significant level set to be p < 0.05. The receiver operating characteristic (ROC) curve was constructed, and the area under the curve (AUC) was calculated to evaluate each parameter's capability in intraoperative hemorrhage subgroup classification. RESULTS Twenty patients had intraoperative hemorrhage (blood loss > 200 mL) during curettage. The hemorrhage group had larger Wash-in, iAUC, Ktrans, Ve, and shorter TTP than the non-hemorrhage group (all P > 0.05). Wash-in had the highest AUC value (0.90), while Ktrans had the lowest value (0.67). Wash-out and Kep were not significantly different between the two groups. CONCLUSION GRASP DCE-MRI has the potential to forecast intraoperative hemorrhage during curettage treatment of CSP, with Wash-in exhibiting the highest predictive performance. This data holds promise for advancing personalized treatment. However, further study is required to compare its effectiveness with other risk factors identified through anatomical MRI and ultrasound.
Collapse
Affiliation(s)
- Zhi-Gang Wang
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No.1617 of Riyue Avenue, Qingyang District, Chengdu, 610091, China
| | - Feng-Leng Yang
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No.1617 of Riyue Avenue, Qingyang District, Chengdu, 610091, China
| | - Chun-Ying Liu
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No.1617 of Riyue Avenue, Qingyang District, Chengdu, 610091, China
| | - Fang Wang
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No.1617 of Riyue Avenue, Qingyang District, Chengdu, 610091, China
| | - Ying Xiong
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiang Zhang
- Department of Gynecology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mei-Ning Chen
- Department of MR Scientific Marketing, Siemens Healthineers, Shanghai, China
| | - Hua Lai
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No.1617 of Riyue Avenue, Qingyang District, Chengdu, 610091, China.
| |
Collapse
|
10
|
Jasik KP, Kleczka A, Franielczyk A. Histopathological Aspects of the Influence of Babesia microti on the Placentas of Infected Female Rats. Vet Sci 2024; 11:18. [PMID: 38250924 PMCID: PMC10819886 DOI: 10.3390/vetsci11010018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/23/2024] Open
Abstract
Babesiosis is perceived mainly an animal disease; however, awareness that Babesia spp. parasites that can cause diseases in humans is increasing significantly. Babesiosis is spread by the bite of an infected tick (Ixodes spp.), but it can also be transmitted by transfusion of infected blood and from an infected mother to her child during pregnancy or childbirth. The parasites multiply in the bloodstream and destroy red blood cells. This study aimed to assess the influence of Babesia microti on the histological structure of the placenta. Histopathological material collected from pregnant rats infected with Babesia microti was used in the experiment. Microscopic images of the placentas were assessed by Mallory staining and by using methylene blue-stained semi-thin sections. In addition, FISH was used to detect parasite DNA. The presence of piroplasms in both maternal and fetal vessels was demonstrated. Babesia microti infection caused vacuolization of syncytioblasts and cytotrophoblasts, accumulation of collagen fibers in placental villi, and increased adhesion of erythrocytes to the vascular walls. These results indicate that Babesia may influence the course of pregnancy and invite further research on the mechanism of piroplasm penetration into cells.
Collapse
Affiliation(s)
- Krzysztof P. Jasik
- Department of Pathology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Ostrogórska 30, 41-200 Sosnowiec, Poland;
| | - Anna Kleczka
- Department of Pathology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Ostrogórska 30, 41-200 Sosnowiec, Poland;
| | | |
Collapse
|
11
|
Zeng S, Liu Z, Yin J, Li S, Jiang M, Yang H, Long Y. Improvement in Clinical Features of L-NAME-Induced Preeclampsia-like Rats through Reduced SERPINA5 Expression. Biomolecules 2023; 13:1792. [PMID: 38136662 PMCID: PMC10742323 DOI: 10.3390/biom13121792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/26/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Pre-eclampsia (PE) is a severe pregnancy disorder that poses a significant health risk to both mother and fetus, with no preventive or therapeutic measures. Our previous research suggested an association between elevated SERPINA5 levels and PE features. This study investigated whether SERPINA5 could be a potential therapeutic target for PE. We established PE-like features in pregnant rats using L-NAME (75 mg/kg/d) treatment. Adenoviruses carrying overexpressed or suppressed SERPINA5 genes were intravenously injected into these PE rats on the fifth and seventh days of pregnancy. We evaluated the rats' systolic blood pressure, urine protein concentration, and placental and fetal metrics and histology. Placental gene expression following SERPINA5 overexpression was evaluated using mRNA sequencing. The L-NAME-induced PE rat model observed a significant increase in placental and peripheral SERPINA5 levels. The overexpression of SERPINA5 exacerbated L-NAME-induced hypertension and proteinuria in pregnant rats. A histology examination revealed a smaller placental junctional zone in L-NAME + overexpressing rats. Placental gene expression analysis in the L-NAME + overexpressing group indicated increased coagulation activation. L-NAME-induced hypertension and proteinuria were mitigated when SERPINA5 expression was suppressed. Additionally, placental development was improved in the SERPINA5-suppressed group. Our findings suggested that SERPINA5 may worsen L-NAME-induced PE-like features by promoting the activation of the coagulation cascade. Therefore, reducing SERPINA5 expression could potentially serve as a therapeutic strategy for PE.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yan Long
- Department of Laboratory, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 510623, China; (S.Z.); (Z.L.); (J.Y.); (S.L.); (M.J.); (H.Y.)
| |
Collapse
|
12
|
Simmers MD, Hudson KM, Baptissart M, Cowley M. Epigenetic control of the imprinted growth regulator Cdkn1c in cadmium-induced placental dysfunction. Epigenetics 2023; 18:2088173. [PMID: 35770551 PMCID: PMC10989690 DOI: 10.1080/15592294.2022.2088173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 05/31/2022] [Indexed: 11/03/2022] Open
Abstract
Cadmium (Cd) is a toxic metal ubiquitous in the environment. In utero, Cd is inefficiently transported to the foetus but causes foetal growth restriction (FGR), likely through impairment of the placenta where Cd accumulates. However, the underlying molecular mechanisms are poorly understood. Cd can modulate the expression of imprinted genes, defined by their transcription from one parental allele, which play critical roles in placental and foetal growth. The expression of imprinted genes is governed by DNA methylation at Imprinting Control Regions (ICRs), which are susceptible to environmental perturbation. The imprinted gene Cdkn1c/CDKN1C is a major regulator of placental development, is implicated in FGR, and shows increased expression in response to Cd exposure in mice. Here, we use a hybrid mouse model of in utero Cd exposure to determine if the increase in placental Cdkn1c expression is caused by changes to ICR DNA methylation and loss of imprinting (LOI). Consistent with prior studies, Cd causes FGR and impacts placental structure and Cdkn1c expression at late gestation. Using polymorphisms to distinguish parental alleles, we demonstrate that increased Cdkn1c expression is not driven by changes to DNA methylation or LOI. We show that Cdkn1c is expressed primarily in the placental labyrinth which is proportionally increased in size in response to Cd. We conclude that the Cd-associated increase in Cdkn1c expression can be fully explained by alterations to placental structure. These results have implications for understanding mechanisms of Cd-induced placental dysfunction and, more broadly, for the study of FGR associated with increased Cdkn1c/CDKN1C expression.
Collapse
Affiliation(s)
- Mark D. Simmers
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NCUSA
| | - Kathleen M. Hudson
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NCUSA
| | - Marine Baptissart
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NCUSA
| | - Michael Cowley
- Center for Human Health and the Environment, and Department of Biological Sciences, North Carolina State University, Raleigh, NCUSA
| |
Collapse
|
13
|
Mineo C, Shaul PW, Bermas BL. The pathogenesis of obstetric APS: a 2023 update. Clin Immunol 2023; 255:109745. [PMID: 37625670 PMCID: PMC11366079 DOI: 10.1016/j.clim.2023.109745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023]
Abstract
The antiphospholipid syndrome (APS) is an autoimmune disease characterized by the persistent presence of antibodies directed against phospholipids and phospholipid-binding proteins that are associated with thrombosis and pregnancy-related morbidity. The latter includes fetal deaths, premature birth and maternal complications. In the early 1990s, a distinct set of autoantibodies, termed collectively antiphospholipid antibodies (aPL), were identified as the causative agents of this disorder. Subsequently histological analyses of the placenta from APS pregnancies revealed various abnormalities, including inflammation at maternal-fetal interface and poor placentation manifested by reduced trophoblast invasion and limited uterine spiral artery remodeling. Further preclinical investigations identified the molecular targets of aPL and the downstream intracellular pathways of key placental cell types. While these discoveries suggest potential therapeutics for this disorder, definitive clinical trials have not been completed. This concise review focuses on the recent developments in the field of basic and translational research pursuing novel mechanisms underlying obstetric APS.
Collapse
Affiliation(s)
- Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States.
| | - Philip W Shaul
- Center for Pulmonary and Vascular Biology, Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States
| | - Bonnie L Bermas
- Division of Rheumatic Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
14
|
Li Q, Li S, Ding J, Pang B, Li R, Cao H, Ling L. MALAT1 modulates trophoblast phenotype via miR-101-3p/VEGFA axis. Arch Biochem Biophys 2023:109692. [PMID: 37437834 DOI: 10.1016/j.abb.2023.109692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/23/2023] [Accepted: 07/09/2023] [Indexed: 07/14/2023]
Abstract
Preeclampsia is a potentially life-threatening condition that can arise due to poor placentation and consequent abnormal uterine spiral artery remodeling. Abnormal placentation, in turn, is associated with aberrant trophoblast cell proliferation and apoptosis. Here, we investigated the lncRNA MALAT1 in trophoblast proliferation during early-onset preeclampsia (ePE). MALAT1 levels were examined in placental tissue samples from ePE patients and control patients. The effects and underlying mechanism of MALAT1 on proliferation, migration, invasion and apoptosis were investigated in the first-trimester extravillous trophoblast HTR-8/SVneo cells and the human choriocarcinoma JAR cells. MALAT1 levels were decreased in the placental tissue samples of ePE patients compared with those of control patients, and the levels of MALAT1 were positively correlated with the neonate birth-weight. Knockdown of MALAT1 attenuated the cell viability, proliferation, migration, invasion and the cell cycle progression of trophoblasts, but promoted the apoptosis of trophoblasts. The MALAT1 knockdown promoted miR-101-3p upregulation and VEGFA downregulation. Inhibitor of miR-101-3p increased vascular endothelial growth factor A (VEGFA) expression, and miR-101-3p mimic inhibited VEGFA expression. Luciferase assays showed that miR-101-3p could bind to both MALAT1 and VEGFA. The MALAT1 knockdown-induced induction in the cell vitality and proliferation were attenuated by miR-101-3p inhibitor. We conclude that endogenous MALAT1 promotes proliferation, migration and invasion of trophoblasts by inhibiting the miR-101-3p expression and the subsequent VEGFA downregulation. The reduced MALAT1 level in placental tissue may be involved in the pathogenesis of the ePE.
Collapse
Affiliation(s)
- Qin Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Shuo Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Jin Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Bo Pang
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Ranran Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Hui Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Li Ling
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241000, China.
| |
Collapse
|
15
|
Massri N, Loia R, Sones JL, Arora R, Douglas NC. Vascular changes in the cycling and early pregnant uterus. JCI Insight 2023; 8:e163422. [PMID: 37288662 PMCID: PMC10393238 DOI: 10.1172/jci.insight.163422] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Uterine vascular remodeling is intrinsic to the cycling and early pregnant endometrium. Maternal regulatory factors such as ovarian hormones, VEGF, angiopoietins, Notch, and uterine natural killer cells significantly mediate these vascular changes. In the absence of pregnancy, changes in uterine vessel morphology and function correlate with different stages of the human menstrual cycle. During early pregnancy, vascular remodeling in rodents and humans results in decreased uterine vascular resistance and increased vascular permeability necessary for pregnancy success. Aberrations in these adaptive vascular processes contribute to increased risk of infertility, abnormal fetal growth, and/or preeclampsia. This Review comprehensively summarizes uterine vascular remodeling in the human menstrual cycle, and in the peri- and post-implantation stages in rodent species (mice and rats).
Collapse
Affiliation(s)
- Noura Massri
- Cell and Molecular Biology Graduate Program and
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Rachel Loia
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Jennifer L. Sones
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Ripla Arora
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
| | - Nataki C. Douglas
- Department of Obstetrics, Gynecology and Reproductive Health and
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| |
Collapse
|
16
|
Simmers MD, Jima DD, Tsuji Y, Cowley M. LncRNA Tuna is activated in cadmium-induced placental insufficiency and drives the NRF2-mediated oxidative stress response. Front Cell Dev Biol 2023; 11:1151108. [PMID: 37325564 PMCID: PMC10267411 DOI: 10.3389/fcell.2023.1151108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/24/2023] [Indexed: 06/17/2023] Open
Abstract
Cadmium (Cd) is a toxic heavy metal found throughout the environment and one of the top ten toxicants of major public health concern identified by the World Health Organization. In utero Cd exposure causes fetal growth restriction, malformation, and spontaneous abortion; however, the mechanisms by which Cd impacts these outcomes are poorly understood. Cd accumulates in the placenta, suggesting that these negative outcomes may be a consequence of disrupted placental function and placental insufficiency. To understand the impact of Cd on gene expression within the placenta, we developed a mouse model of Cd-induced fetal growth restriction through maternal consumption of CdCl2 and performed RNA-seq on control and CdCl2 exposed placentae. The top differentially expressed transcript was the Tcl1 Upstream Neuron-Associated (Tuna) long non-coding RNA, which was upregulated over 25-fold in CdCl2 exposed placentae. Tuna has been shown to be critical for neural stem cell differentiation. However, within the placenta, there is no evidence that Tuna is normally expressed or functional at any developmental stage. To determine the spatial expression of Cd-activated Tuna within the placenta, we used in situ hybridization as well as placental layer-specific RNA isolation and analysis. Both methods confirmed the absence of Tuna expression in control samples and determined that Cd-induced Tuna expression is specific to the junctional zone. Since many lncRNAs regulate gene expression, we hypothesized that Tuna forms part of the mechanism of Cd-induced transcriptomic changes. To test this, we over-expressed Tuna in cultured choriocarcinoma cells and compared gene expression profiles to those of control and CdCl2 exposed cells. We demonstrate significant overlap between genes activated by Tuna overexpression and genes activated by CdCl2 exposure, with enrichment in the NRF2-mediated oxidative stress response. Herein we analyze the NRF2 pathway and show that Tuna increases NRF2/NRF2 both at the transcript and protein levels. Tuna drives increased NRF2 target gene expression, a result that is abrogated with the use of an NRF2 inhibitor, confirming that Tuna activates oxidative stress response genes through this pathway. This work identifies the lncRNA Tuna as a potential novel player in Cd-induced placental insufficiency.
Collapse
Affiliation(s)
- Mark D. Simmers
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Dereje D. Jima
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States
| | - Yoshiaki Tsuji
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Michael Cowley
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
17
|
Kanter J, Gordon SM, Mani S, Sokalska A, Park JY, Senapati S, Huh DD, Mainigi M. Hormonal stimulation reduces numbers and impairs function of human uterine natural killer cells during implantation. Hum Reprod 2023; 38:1047-1059. [PMID: 37075311 PMCID: PMC10501469 DOI: 10.1093/humrep/dead069] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/05/2023] [Indexed: 04/21/2023] Open
Abstract
STUDY QUESTION How does an altered maternal hormonal environment, such as that seen during superovulation with gonadotropins in ART, impact human uterine immune cell distribution and function during the window of implantation? SUMMARY ANSWER Hormonal stimulation with gonadotropins alters abundance of maternal immune cells including uterine natural killer (uNK) cells and reduces uNK cell ability to promote extravillous trophoblast (EVT) invasion. WHAT IS KNOWN ALREADY An altered maternal hormonal environment, seen following ART, can lead to increased risk for adverse perinatal outcomes associated with disordered placentation. Maternal immune cells play an essential role in invasion of EVTs, a process required for proper establishment of the placenta, and adverse perinatal outcomes have been associated with altered immune cell populations. How ART impacts maternal immune cells and whether this can in turn affect implantation and placentation in humans remain unknown. STUDY DESIGN, SIZE, DURATION A prospective cohort study was carried out between 2018 and 2021 on 51 subjects: 20 from natural cycles 8 days after LH surge; and 31 from stimulated IVF cycles 7 days after egg retrieval. PARTICIPANTS/MATERIALS, SETTING, METHODS Endometrial biopsies and peripheral blood samples were collected during the window of implantation in subjects with regular menstrual cycles or undergoing superovulation. Serum estradiol and progesterone levels were measured by chemiluminescent competitive immunoassay. Immune cell populations in blood and endometrium were analyzed using flow cytometry. uNK cells were purified using fluorescence-activated cell sorting and were subjected to RNA sequencing (RNA-seq). Functional changes in uNK cells due to hormonal stimulation were evaluated using the implantation-on-a-chip (IOC) device, a novel bioengineered platform using human primary cells that mimics early processes that occur during pregnancy in a physiologically relevant manner. Unpaired t-tests, one-way ANOVA, and pairwise multiple comparison tests were used to statistically evaluate differences. MAIN RESULTS AND THE ROLE OF CHANCE Baseline characteristics were comparable for both groups. As expected, serum estradiol levels on the day of biopsy were significantly higher in stimulated (superovulated) patients (P = 0.0005). In the setting of superovulation, we found an endometrium-specific reduction in the density of bulk CD56+ uNK cells (P < 0.05), as well as in the uNK3 subpopulation (P = 0.025) specifically (CD103+ NK cells). In stimulated samples, we also found that the proportion of endometrial B cells was increased (P < 0.0001). Our findings were specific to the endometrium and not seen in peripheral blood. On the IOC device, uNK cells from naturally cycling secretory endometrium promote EVT invasion (P = 0.03). However, uNK cells from hormonally stimulated endometrium were unable to significantly promote EVT invasion, as measured by area of invasion, depth of invasion, and number of invaded EVTs by area. Bulk RNA-seq of sorted uNK cells from stimulated and unstimulated endometrium revealed changes in signaling pathways associated with immune cell trafficking/movement and inflammation. LIMITATIONS, REASONS FOR CAUTION Patient numbers utilized for the study were low but were enough to identify significant overall population differences in select immune cell types. With additional power and deeper immune phenotyping, we may detect additional differences in immune cell composition of blood and endometrium in the setting of hormonal stimulation. Flow cytometry was performed on targeted immune cell populations that have shown involvement in early pregnancy. A more unbiased approach might identify changes in novel maternal immune cells not investigated in this study. We performed RNA-seq only on uNK cells, which demonstrated differences in gene expression. Ovarian stimulation may also impact gene expression and function of other subsets of immune cells, as well as other cell types within the endometrium. Finally, the IOC device, while a major improvement over existing in vitro methods to study early pregnancy, does not include all possible maternal cells present during early pregnancy, which could impact functional effects seen. Immune cells other than uNK cells may impact invasion of EVTs in vitro and in vivo, though these remain to be tested. WIDER IMPLICATIONS OF THE FINDINGS These findings demonstrate that hormonal stimulation affects the distribution of uNK cells during the implantation window and reduces the proinvasive effects of uNK cells during early pregnancy. Our results provide a potential mechanism by which fresh IVF cycles may increase risk of disorders of placentation, previously linked to adverse perinatal outcomes. STUDY FUNDING/COMPETING INTEREST(S) Research reported in this publication was supported by the University of Pennsylvania University Research Funding (to M.M.), the Eunice Kennedy Shriver National Institute of Child Health and Human Development (P50HD068157 to M.M., S.S., and S.M.), National Center for Advancing Translational Sciences of the National Institutes of Health (TL1TR001880 to J.K.), the Institute for Translational Medicine and Therapeutics of the Perelman School of Medicine at the University of Pennsylvania, the Children's Hospital of Philadelphia Research Institute (to S.M.G.), and the National Institute of Allergy and Infectious Diseases (K08AI151265 to S.M.G.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- J Kanter
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - S M Gordon
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - S Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - A Sokalska
- Division of Reproductive Endocrinology and Infertility, Stanford University, Stanford, CA, USA
| | - J Y Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - S Senapati
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - D D Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - M Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
18
|
Ma Y, Hu Y, Ma J. Animal models of the placenta accreta spectrum: current status and further perspectives. Front Endocrinol (Lausanne) 2023; 14:1118168. [PMID: 37223034 PMCID: PMC10200980 DOI: 10.3389/fendo.2023.1118168] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 03/20/2023] [Indexed: 05/25/2023] Open
Abstract
Placenta accreta spectrum disorder (PAS) is a kind of disease of placentation defined as abnormal trophoblast invasion of part or all of the placenta into the myometrium, even penetrating the uterus. Decidual deficiency, abnormal vascular remodeling in the maternal-fetal interface, and excessive invasion by extravillous trophoblast (EVT) cells contribute to its onset. However, the mechanisms and signaling pathways underlying such phenotypes are not fully understood, partly due to the lack of suitable experimental animal models. Appropriate animal models will facilitate the comprehensive and systematic elucidation of the pathogenesis of PAS. Due to the remarkably similar functional placental villous units and hemochorial placentation to humans, the current animal models of PAS are based on mice. There are various mouse models induced by uterine surgery to simulate different phenotypes of PAS, such as excessive invasion of EVT or immune disturbance at the maternal-fetal interface, which could define the pathological mechanism of PAS from the perspective of the "soil." Additionally, genetically modified mouse models could be used to study PAS, which is helpful to exploring the pathogenesis of PAS from the perspectives of both "soil" and "seed," respectively. This review details early placental development in mice, with a focus on the approaches of PAS modeling. Additionally, the strengths, limitations and the applicability of each strategy and further perspectives are summarized to provide the theoretical foundation for researchers to select appropriate animal models for various research purposes. This will help better determine the pathogenesis of PAS and even promote possible therapy.
Collapse
Affiliation(s)
- Yongdan Ma
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Yongyan Hu
- Laboratory Animal Center, Peking University First Hospital, Beijing, China
| | - Jingmei Ma
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
- Beijing Key Laboratory of Maternal Fetal Medicine of Gestational Diabetes Mellitus, Beijing, China
| |
Collapse
|
19
|
Santos LC, de Souza CA, Silva JF, Ocarino NM, Serakides R. Maternal hyperthyroidism alters the immunological mediators profile and population of natural killers cells in decidua of rats. Acta Histochem 2023; 125:152026. [PMID: 37058857 DOI: 10.1016/j.acthis.2023.152026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023]
Abstract
Decidual immunological mediators modulate placental formation, decidualization and fetal development. However, the effect of maternal hyperthyroidism on decidual immunology needs further research. The aim of this study was to evaluate the population of uterine natural killer cells (uNKs) and the expression of immunological mediators in the decidua of female rats throughout pregnancy. Wistar rats were used and hyperthyroidism was induced by daily administration of L-thyroxine (T4) throughout pregnancy. The population of uNK cells in decidua was evaluated by immunostaining Lectin DBA, as well as the expression of interferon γ (INFγ), macrophage migration inhibitory factor (MIF), interleukin 15 (IL-15) and inducible nitric oxide synthase (iNOS) at 7, 10, 12, 14 and 19 days of gestation (DG). Maternal hyperthyroidism reduced the DBA+ uNK cell population in the decidua at 7 (P < 0.05) and 10 (P < 0.01) DGs compared to that in the control group, while it increased in the basal decidua (P < 0.05) and metrial gland (P < 0.0001) at the 12th DG. Hyperthyroidism also increased immunostaining of IL-15 (P < 0.0001), INFγ (P < 0.05), and MIF (P < 0.05) in the 7th DG, and increased immunostaining of IL-15 (P < 0.0001) and MIF (P < 0.01) in the 10th DG. However, excess thyroxine reduced IL-15 expression in the metrial gland and/or basal decidua in the 12th (P < 0.05), 14th (P < 0.01), and 19th (P < 0.001) DGs, as was also observed for INFγ in the basal decidua (P<0.001) and metrial gland (P < 0.0001) in the 12th DG. Regarding iNOS, an antiinflammatory cytokine, lower expression was observed in the basal decidua of hyperthyroid animals at 7 and 12 DGs (P < 0.05), whereas an increase occurred in the 10th DG (P < 0.05). These data demonstrate that maternal hyperthyroidism in female rats, particularly between 7 and 10 DGs, reduces the population of DBA+ uNKs in the decidua and increases the expression of inflammatory cytokines, suggesting a more proinflammatory environment in early pregnancy caused by this gestational disease.
Collapse
Affiliation(s)
- Luciano Cardoso Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900 Ilheus, Brazil
| | - Cíntia Almeida de Souza
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900 Ilheus, Brazil
| | - Natália Melo Ocarino
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil
| | - Rogéria Serakides
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, Brazil.
| |
Collapse
|
20
|
Waker CA, Hwang AE, Bowman-Gibson S, Chandiramani CH, Linkous B, Stone ML, Keoni CI, Kaufman MR, Brown TL. Mouse models of preeclampsia with preexisting comorbidities. Front Physiol 2023; 14:1137058. [PMID: 37089425 PMCID: PMC10117893 DOI: 10.3389/fphys.2023.1137058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/17/2023] [Indexed: 04/08/2023] Open
Abstract
Preeclampsia is a pregnancy-specific condition and a leading cause of maternal and fetal morbidity and mortality. It is thought to occur due to abnormal placental development or dysfunction, because the only known cure is delivery of the placenta. Several clinical risk factors are associated with an increased incidence of preeclampsia including chronic hypertension, diabetes, autoimmune conditions, kidney disease, and obesity. How these comorbidities intersect with preeclamptic etiology, however, is not well understood. This may be due to the limited number of animal models as well as the paucity of studies investigating the impact of these comorbidities. This review examines the current mouse models of chronic hypertension, pregestational diabetes, and obesity that subsequently develop preeclampsia-like symptoms and discusses how closely these models recapitulate the human condition. Finally, we propose an avenue to expand the development of mouse models of preeclampsia superimposed on chronic comorbidities to provide a strong foundation needed for preclinical testing.
Collapse
Affiliation(s)
- Christopher A. Waker
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Amy E. Hwang
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Scout Bowman-Gibson
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Chandni H. Chandiramani
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Bryce Linkous
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Madison L. Stone
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Chanel I. Keoni
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Melissa R. Kaufman
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
| | - Thomas L. Brown
- Department of Neuroscience, Cell Biology and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
- Department of Obstetrics and Gynecology, Boonshoft School of Medicine, Wright State University, Dayton, OH, United States
- *Correspondence: Thomas L. Brown,
| |
Collapse
|
21
|
Wu Z, Hu G, Gong T, Hu Q, Hong L, Zhang Y, Ao Z. RACK1 may participate in placental development at mid-gestation via regulating trophoblast cell proliferation and migration in pigs. Mol Reprod Dev 2023; 90:248-259. [PMID: 36916007 DOI: 10.1002/mrd.23680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/15/2023]
Abstract
Intrauterine growth restriction (IUGR) is a severe complication in swine production. Placental insufficiency is responsible for inadequate fetal growth, but the specific etiology of placental dysfunction-induced IUGR in pigs remains poorly understood. In this work, placenta samples supplying the lightest weight (LW) and mean weight (MW) pig fetuses in the litter at Day 65 (D65) of gestation were collected, and the relationship between fetal growth and placental morphologies and functions was investigated using histomorphological analysis, RNA sequencing, quantitative polymerase chain reaction, and in vitro experiment in LW and MW placentas. Results showed that the folded structure of the epithelial bilayer of LW placentas followed a poor and incomplete development compared with that of MW placentas. A total of 654 differentially expressed genes (DEGs) were screened out between the LW and MW placentas, and the gene encodes receptor for activated C kinase 1 (RACK1) was found to be downregulated in LW placentas. The DEGs were mainly enriched in translation, ribosome, protein synthesis, and mammalian target of rapamycin (mTOR) signaling pathway according to gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. In vitro experiments indicated that the decreased RACK1 in LW placentas may be involved in abnormal development of placental folds (PFs) by inhibiting the proliferation and migration of porcine trophoblast cells. Taken together, these results revealed that RACK1 may be a vital regulator in the development of PFs via regulating trophoblast cell proliferation and migration in pigs.
Collapse
Affiliation(s)
- Zhimin Wu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China.,Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, China
| | - Guangling Hu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China.,Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, China
| | - Ting Gong
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China.,Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, China
| | - Qun Hu
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Linjun Hong
- National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yiyu Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China.,Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, China
| | - Zheng Ao
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China.,Guizhou Provincial Key Laboratory of Animal Genetics, Breeding and Reproduction, College of Animal Science, Guizhou University, Guiyang, China
| |
Collapse
|
22
|
Meakin C, Kim C, Lampert T, Aleksunes LM. High-throughput screening of toxicants that modulate extravillous trophoblast migration. Toxicol Lett 2023; 375:1-7. [PMID: 36535517 PMCID: PMC9877196 DOI: 10.1016/j.toxlet.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Migration and subsequent invasion of extravillous trophoblasts into the uterus is essential for proper formation of the placenta. Disruption of these processes may result in poor pregnancy outcomes including preeclampsia, placenta accreta, fetal growth restriction, or fetal death. Currently, there are several methods for quantifying cell migration and invasion in vitro, each with limitations. Therefore, we developed a novel, high-throughput method to screen chemicals for their ability to alter human trophoblast migration. Human HTR8/SVneo trophoblast cells were cultured in Oris™ cell migration plates containing stopper barriers. After EVT cells attached and chemicals were added to media, stoppers were removed thereby creating a cell-free detection zone for migration. Entry of trophoblasts into this zone was monitored through imaging every 6 h and used to calculate a relative cell density. Chemicals known to increase (epidermal growth factor) and decrease (pertussis toxin and cadmium) trophoblast migration were used to validate this in vitro method. Next, a panel of environmental chemicals including bisphenols, mycoestrogens, and flame retardants, were screened for their ability to alter trophoblast invasion. In conclusion, a real-time method to track extravillous trophoblast migration offers potential for screening contaminants as placental toxicants.
Collapse
Affiliation(s)
- Cassandra Meakin
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Christine Kim
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | | | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA; Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
23
|
Vu HT, Kaur H, Kies KR, Starks RR, Tuteja G. Identifying novel regulators of placental development using time-series transcriptome data. Life Sci Alliance 2023; 6:6/2/e202201788. [PMID: 36622342 PMCID: PMC9748866 DOI: 10.26508/lsa.202201788] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/15/2022] Open
Abstract
The placenta serves as a connection between the mother and the fetus during pregnancy, providing the fetus with oxygen, nutrients, and growth hormones. However, the regulatory mechanisms and dynamic gene interaction networks underlying early placental development are understudied. Here, we generated RNA-sequencing data from mouse fetal placenta at embryonic days 7.5, 8.5, and 9.5 to identify genes with timepoint-specific expression, then inferred gene interaction networks to analyze highly connected network modules. We determined that timepoint-specific gene network modules were associated with distinct developmental processes, and with similar expression profiles to specific human placental cell populations. From each module, we identified hub genes and their direct neighboring genes, which were predicted to govern placental functions. We confirmed that four novel candidate regulators identified through our analyses regulate cell migration in the HTR-8/SVneo cell line. Overall, we predicted several novel regulators of placental development expressed in specific placental cell types using network analysis of bulk RNA-sequencing data. Our findings and analysis approaches will be valuable for future studies investigating the transcriptional landscape of early development.
Collapse
Affiliation(s)
- Ha Th Vu
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA.,Bioinformatics and Computational Biology, Iowa State University, Ames, IA, USA
| | - Haninder Kaur
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA
| | - Kelby R Kies
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA.,Bioinformatics and Computational Biology, Iowa State University, Ames, IA, USA
| | - Rebekah R Starks
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA.,Bioinformatics and Computational Biology, Iowa State University, Ames, IA, USA
| | - Geetu Tuteja
- Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, USA .,Bioinformatics and Computational Biology, Iowa State University, Ames, IA, USA
| |
Collapse
|
24
|
Arutjunyan AV, Kerkeshko GO, Milyutina YP, Shcherbitskaia AD, Zalozniaia IV, Mikhel AV, Inozemtseva DB, Vasilev DS, Kovalenko AA, Kogan IY. Imbalance of Angiogenic and Growth Factors in Placenta in Maternal Hyperhomocysteinemia. BIOCHEMISTRY (MOSCOW) 2023; 88:262-279. [PMID: 37072327 DOI: 10.1134/s0006297923020098] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Numerous studies have shown that various adverse factors of different nature and action mechanisms have similar negative influence on placental angiogenesis, resulting in insufficiency of placental blood supply. One of the risk factors for pregnancy complications with placental etiology is an increased level of homocysteine in the blood of pregnant women. However, the effect of hyperhomocysteinemia (HHcy) on the development of the placenta and, in particular, on the formation of its vascular network is at present poorly understood. The aim of this work was to study the effect of maternal HHcy on the expression of angiogenic and growth factors (VEGF-A, MMP-2, VEGF-B, BDNF, NGF), as well as their receptors (VEGFR-2, TrkB, p75NTR), in the rat placenta. The effects of HHcy were studied in the morphologically and functionally different maternal and fetal parts of the placenta on the 14th and 20th day of pregnancy. The maternal HHcy caused increase in the levels of oxidative stress and apoptosis markers accompanied by an imbalance of the studied angiogenic and growth factors in the maternal and/or fetal part of the placenta. The influence of maternal HHcy in most cases manifested in a decrease in the protein content (VEGF-A), enzymatic activity (MMP-2), gene expression (VEGFB, NGF, TRKB), and accumulation of precursor form (proBDNF) of the investigated factors. In some cases, the effects of HHcy differed depending on the placental part and stage of development. The influence of maternal HHcy on signaling pathways and processes controlled by the studied angiogenic and growth factors could lead to incomplete development of the placental vasculature and decrease in the placental transport, resulting in fetal growth restriction and impaired fetal brain development.
Collapse
Affiliation(s)
- Alexander V Arutjunyan
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia.
| | - Gleb O Kerkeshko
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Yulia P Milyutina
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
- St. Petersburg State Pediatric Medical University, Russian Ministry of Health, St. Petersburg, 194100, Russia
| | - Anastasiia D Shcherbitskaia
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - Irina V Zalozniaia
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Anastasiia V Mikhel
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Daria B Inozemtseva
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| | - Dmitrii S Vasilev
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - Anna A Kovalenko
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, 194223, Russia
| | - Igor Yu Kogan
- D. O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, St. Petersburg, 199034, Russia
| |
Collapse
|
25
|
Long Y, Zeng S, Gao F, Liu F, Zhang Y, Zhou C, Zhu C, Zhao X, Han M, Gan Q, Ye W, Zeng F, Song C, Jiang M, Lash GE, Yang H. SERPINA5 may promote the development of preeclampsia by disruption of the uPA/uPAR pathway. Transl Res 2023; 251:14-26. [PMID: 35717024 DOI: 10.1016/j.trsl.2022.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/24/2022] [Accepted: 06/10/2022] [Indexed: 12/24/2022]
Abstract
Preeclampsia (PE) is the leading cause of maternal and fetal morbidity or mortality but lacks reliable methods for early diagnosis. In a previous study, serum SERPINA5 levels were higher in women with PE before the clinical manifestation of the disease. This study aimed to evaluate the efficacy of SERPINA5 in predicting PE and investigate its role in trophoblast cell biology. A multicenter, 2-stage observational case-control study was performed to develop and validate an early predictive PE model based on SERPINA5, maternal characteristics, and inflammatory factors. To further understand the relationship between SERPINA5 and PE, SERPINA5 was overexpressed or knocked down in extravillous trophoblast cells (EVT) and a pregnant rat model. After development and initial validation, a model that combined SERPINA5 and inflammatory factors had a high predictive ability for PE before 20 weeks gestation with an AUC of 0.90 (95% CI 0.83-0.96). It also demonstrated that SERPINA5 inhibited primary EVT cell invasion by disrupting the urokinase-type plasminogen activator/urokinase-type plasminogen activator receptor (uPA/uPAR) pathway, in turn, is involved in the development of PE. In vivo experiments also proved that overexpression of SERPINA5 induced a PE-like syndrome (hypertension and proteinuria) in pregnant rats. Therefore, serum SERPINA5 is a promising early biomarker of PE, suggesting that it may be involved in placental development through its action on the uPA/uPAR system prior to the clinical manifestation of PE.
Collapse
Affiliation(s)
- Yan Long
- Department of Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Shanshui Zeng
- Department of Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Fei Gao
- Department of Laboratory Medicine Center, Nanfang Hospital, Southern Medical University/The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Fei Liu
- Department of Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yonggang Zhang
- Department of Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen, China
| | - Cheng Zhou
- Laboratory of Molecular Diagnostics, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Chunyan Zhu
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Xueqin Zhao
- Department of Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Mengru Han
- Department of Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qiangsheng Gan
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Weitao Ye
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Fangling Zeng
- Department of Gynaecology and Obstetrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China
| | - Chunlin Song
- Laboratory of Molecular Diagnostics, Southern Medical University Affiliated Maternal & Child Health Hospital of Foshan, Foshan, China
| | - Min Jiang
- Department of Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| | - Gendie E Lash
- Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, China.
| | - Hongling Yang
- Department of Laboratory, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
26
|
Liang L, Chen Y, Wu C, Cao Z, Xia L, Meng J, He L, Yang C, Wang Z. MicroRNAs: key regulators of the trophoblast function in pregnancy disorders. J Assist Reprod Genet 2023; 40:3-17. [PMID: 36508034 PMCID: PMC9742672 DOI: 10.1007/s10815-022-02677-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
The placenta is essential for a successful pregnancy and healthy intrauterine development in mammals. During human pregnancy, the growth and development of the placenta are inseparable from the rapid proliferation, invasion, and migration of trophoblast cells. Previous reports have shown that the occurrence of many pregnancy disorders may be closely related to the dysfunction of trophoblasts. However, the function regulation of human trophoblast cells in the placenta is poorly understood. Therefore, studying the factors that regulate the function of trophoblast cells is necessary. MicroRNAs (miRNAs) are small, non-coding, single-stranded RNA molecules. Increasing evidence suggests that miRNAs play a crucial role in regulating trophoblast functions. This review outlines the role of miRNAs in regulating the function of trophoblast cells and several common signaling pathways related to miRNA regulation in pregnancy disorders.
Collapse
Affiliation(s)
- Lingli Liang
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| | - Yanjun Chen
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| | - Chunyan Wu
- grid.412017.10000 0001 0266 8918Department of Cardiovascular, The Third Affiliated Hospital of University of South China, Hengyang, 421001 China
| | - Zitong Cao
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| | - Linzhen Xia
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| | - Jun Meng
- grid.461579.8Department of Function, The First Affiliated Hospital of University of South China, Hengyang, 421001 China
| | - Lu He
- grid.461579.8Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang, 421001 China
| | - Chunfen Yang
- grid.461579.8Department of Gynecology, The First Affiliated Hospital of University of South China, Hengyang, 421001 China
| | - Zuo Wang
- grid.412017.10000 0001 0266 8918Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, Hengyang, 421001 China
| |
Collapse
|
27
|
Cui K, Chen T, Zhu Y, Shi Y, Guo Y, Qin J. Engineering placenta-like organoids containing endogenous vascular cells from human-induced pluripotent stem cells. Bioeng Transl Med 2023; 8:e10390. [PMID: 36684087 PMCID: PMC9842056 DOI: 10.1002/btm2.10390] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/17/2022] [Accepted: 07/27/2022] [Indexed: 01/25/2023] Open
Abstract
The placenta is an essential organ that maintains the health of both the fetus and its mother. Understanding the development of human placenta has been hindered by the limitations of existing animal models and monolayer cell cultures. Models that can recapitulate the essential aspects of human placental multicellular components and vasculature are still lacking. Herein, we presented a new strategy to establish placenta-like organoids with vascular-like structures from human-induced pluripotent stem cells in a defined three-dimensional (3D) culture system. The resulting placenta-like tissue resembles first-trimester human placental development in terms of complex placental components and secretory function. The multicellular tissue was characterized by the inclusion of trophoblasts (cytotrophoblasts, syncytiotrophoblasts, extravillous trophoblasts, and other endogenous vascular cells), which were identified by immunofluorescence, flow cytometry analyses, real-time quantitative reverse transcription polymerase chain reaction and single-cell RNA-seq. Moreover, the 3D tissue was able to secrete the placenta-specific hormone human chorionic gonadotropin β (hCG-β) and vascular endothelial growth factor A (VEGFA). The tissue responded to the inflammatory factor tumor necrosis factor-α (TNF-α) and VEGF receptor inhibitors. This new model system can represent the major features of placental cellular components, and function, which have not been realized in 2D monolayer cultures. The developed tissue system might open new avenues for studying normal early human placental development and its disease states.
Collapse
Affiliation(s)
- Kangli Cui
- Division of Biotechnology, CAS Key Laboratory of SSACDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Tingwei Chen
- Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Yujuan Zhu
- Division of Biotechnology, CAS Key Laboratory of SSACDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yang Shi
- Dalian Municipal Woman and Children's Medical CenterDalianChina
| | - Yaqiong Guo
- Division of Biotechnology, CAS Key Laboratory of SSACDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
| | - Jianhua Qin
- Division of Biotechnology, CAS Key Laboratory of SSACDalian Institute of Chemical Physics, Chinese Academy of SciencesDalianChina
- University of Chinese Academy of SciencesBeijingChina
- Beijing Institute for Stem Cell and RegenerationBeijingChina
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
28
|
Ko G, Jeon TJ, Kim SM. Trophoblast Migration with Different Oxygen Levels in a Gel-Patterned Microfluidic System. MICROMACHINES 2022; 13:2216. [PMID: 36557515 PMCID: PMC9783522 DOI: 10.3390/mi13122216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 06/17/2023]
Abstract
In the placenta, substances such as nutrients, oxygen, and by-products are exchanged between the mother and the fetus, and the proper formation of the placenta determines the success of pregnancy, including the growth of the fetus. Preeclampsia is an obstetric disease in which the incomplete formation of the placenta occurs, which is known to occur when there is an abnormality in the invasion of trophoblast cells. The invasion of trophoblast cells is controlled by oxygen concentration, and HIF-1α changes according to oxygen concentration, showing a difference in cell mobility. MMP-2 and MMP-9 are observed to be high in the endometrium involved in trophoblast invasion, and the expression is regulated according to the oxygen concentration. In this experiment, cell culture was conducted using a gel-patterned system with a hypoxic chamber. Before the chip experiment, the difference in the expression of MMP-2 and MMP-9 according to the oxygen concentration was confirmed using a hypoxia chamber. After that, trophoblast cells (HTR8/SVneo) and endothelial cells (HUVECs) were separated and cultured through a physical barrier through a hydrogel on a microfluidic chip. Cells were cultured in a hypoxic chamber under controlled oxygen levels. It was confirmed that the mobility of trophoblast cells in culture on the chip was upregulated in a hypoxic environment through oxygen control. This suggests that the formation of a hypoxic environment in the endometrium where the invasion of trophoblast cells occurs plays a role in increasing cell mobility.
Collapse
Affiliation(s)
- Gun Ko
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Tae-Joon Jeon
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Sun Min Kim
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Department of Mechanical Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
29
|
Attenuated effect of zinc gluconate on oxidative stress, inflammation, and angiogenic imbalance in pre-eclampsia rats. Life Sci 2022; 310:121055. [DOI: 10.1016/j.lfs.2022.121055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/29/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
|
30
|
Reliability of Rodent and Rabbit Models in Preeclampsia Research. Int J Mol Sci 2022; 23:ijms232214344. [PMID: 36430816 PMCID: PMC9696504 DOI: 10.3390/ijms232214344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
In vivo studies on the pathology of gestation, including preeclampsia, often use small mammals such as rabbits or rodents, i.e., mice, rats, hamsters, and guinea pigs. The key advantage of these animals is their short reproductive cycle; in addition, similar to humans, they also develop a haemochorial placenta and present a similar transformation of maternal spiral arteries. Interestingly, pregnant dams also demonstrate a similar reaction to inflammatory factors and placentally derived antiangiogenic factors, i.e., soluble fms-like tyrosine kinase 1 (sFlt-1) or soluble endoglin-1 (sEng), as preeclamptic women: all animals present an increase in blood pressure and usually proteinuria. These constitute the classical duet that allows for the recognition of preeclampsia. However, the time of initiation of maternal vessel remodelling and the depth of trophoblast invasion differs between rabbits, rodents, and humans. Unfortunately, at present, no known animal replicates a human pregnancy exactly, and hence, the use of rabbit and rodent models is restricted to the investigation of individual aspects of human gestation only. This article compares the process of placentation in rodents, rabbits, and humans, which should be considered when planning experiments on preeclampsia; these aspects might determine the success, or failure, of the study. The report also reviews the rodent and rabbit models used to investigate certain aspects of the pathomechanism of human preeclampsia, especially those related to incorrect trophoblast invasion, placental hypoxia, inflammation, or maternal endothelial dysfunction.
Collapse
|
31
|
Buss G, Stratton MB, Milenkovic L, Stearns T. Postmitotic centriole disengagement and maturation leads to centrosome amplification in polyploid trophoblast giant cells. Mol Biol Cell 2022; 33:ar118. [PMID: 36001376 PMCID: PMC9634975 DOI: 10.1091/mbc.e22-05-0182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
DNA replication is normally coupled with centriole duplication in the cell cycle. Trophoblast giant cells (TGCs) of the placenta undergo endocycles resulting in polyploidy but their centriole state is not known. We used a cell culture model for TGC differentiation to examine centriole and centrosome number and properties. Before differentiation, trophoblast stem cells (TSCs) have either two centrioles before duplication or four centrioles after. We find that the average nuclear area increases approximately eight-fold over differentiation, but most TGCs do not have more than four centrioles. However, these centrioles become disengaged, acquire centrosome proteins, and can nucleate microtubules. In addition, some TGCs undergo further duplication and disengagement of centrioles, resulting in substantially higher numbers. Live imaging revealed that disengagement and separation are centriole autonomous and can occur asynchronously. Centriole amplification, when present, occurs by the standard mechanism of one centriole generating one procentriole. PLK4 inhibition blocks centriole formation in differentiating TGCs but does not affect endocycle progression. In summary, centrioles in TGC endocycles undergo disengagement and conversion to centrosomes. This increases centrosome number but to a limited extent compared with DNA reduplication.
Collapse
Affiliation(s)
- Garrison Buss
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305
| | | | | | - Tim Stearns
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305,Department of Biology, Stanford University, Stanford, CA 94305,*Address correspondence to: Tim Stearns ()
| |
Collapse
|
32
|
Dos Anjos Cordeiro JM, Santos LC, de Oliveira LS, Santos BR, Santos EO, Barbosa EM, de Macêdo IO, de Freitas GJC, Santos DDA, de Lavor MSL, Silva JF. Maternal hypothyroidism causes oxidative stress and endoplasmic reticulum stress in the maternal-fetal interface of rats. Free Radic Biol Med 2022; 191:24-39. [PMID: 36038036 DOI: 10.1016/j.freeradbiomed.2022.08.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 08/02/2022] [Accepted: 08/23/2022] [Indexed: 01/11/2023]
Abstract
Maternal hypothyroidism is associated with pre-eclampsia and intrauterine growth restriction, gestational diseases involving oxidative stress (OS) and endoplasmic reticulum stress (ERS) in the placenta. However, it is not known whether hypothyroidism also causes OS and ERS at the maternal-fetal interface. The aim was to evaluate the fetal-placental development and the expression of mediators of OS and of the unfolded protein response (UPR) in the maternal-fetal interface of hypothyroid rats. Hypothyroidism was induced in Wistar rats with propylthiouracil and the fetal-placental development and placental and decidual expression of antioxidant, hypoxia, and UPR mediators were analyzed at 14 and 18 days of gestation (DG), as well the expression of 8-OHdG and MDA, and reactive oxygen species (ROS) and peroxynitrite levels. Hypothyroidism reduced fetal weight at 14 and 18 DG, in addition to increasing the percentage of fetal death and reducing the weight of the uteroplacental unit at 18 DG. At 14 DG, there was greater decidual and/or placental immunostaining of Hif1α, 8-OHdG, MDA, SOD1, GPx1/2, Grp78 and CHOP in hypothyroid rats, while there was a reduction in placental and/or decidual gene expression of Sod1, Gpx1, Atf6, Perk, Ho1, Xbp1, Grp78 and Chop in the same gestational period. At 18 DG, hypothyroidism increased the placental ROS levels and the decidual and/or placental immunostaining of HIF1α, 8-OHdG, MDA, ATF4, GRP78 and CHOP, while it reduced the immunostaining and enzymatic activity of SOD1, CAT, GST. Hypothyroidism increased the placental mRNA expression of Hifα, Nrf2, Sod2, Gpx1, Cat, Perk, Atf6 and Chop at 18 DG, while decreasing the decidual expression of Sod2, Cat and Atf6. These findings demonstrated that fetal-placental restriction in female rats with hypothyroidism is associated with hypoxia and dysregulation in placental and decidual expression of UPR mediators and antioxidant enzymes, and activation of oxidative stress and endoplasmic reticulum stress at the maternal-fetal interface.
Collapse
Affiliation(s)
- Jeane Martinha Dos Anjos Cordeiro
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus SoaneNazare de Andrade, 45662-900, Ilheus, Brazil
| | - Luciano Cardoso Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus SoaneNazare de Andrade, 45662-900, Ilheus, Brazil
| | - Luciana Santos de Oliveira
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus SoaneNazare de Andrade, 45662-900, Ilheus, Brazil
| | - Bianca Reis Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus SoaneNazare de Andrade, 45662-900, Ilheus, Brazil
| | - Emilly Oliveira Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus SoaneNazare de Andrade, 45662-900, Ilheus, Brazil
| | - Erikles Macêdo Barbosa
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus SoaneNazare de Andrade, 45662-900, Ilheus, Brazil
| | - Isabela Oliveira de Macêdo
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus SoaneNazare de Andrade, 45662-900, Ilheus, Brazil
| | - Gustavo José Cota de Freitas
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniel de Assis Santos
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mário Sérgio Lima de Lavor
- Hospital Veterinario, Departamento de Ciencias Agrarias e Ambientais, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus SoaneNazare de Andrade, 45662-900, Ilheus, Brazil.
| |
Collapse
|
33
|
Rusidzé M, Faure MC, Sicard P, Raymond-Letron I, Giton F, Vessieres E, Prevot V, Henrion D, Arnal JF, Cornil CA, Lenfant F. Loss of function of the maternal membrane oestrogen receptor ERα alters expansion of trophoblast cells and impacts mouse fertility. Development 2022; 149:dev200683. [PMID: 36239412 PMCID: PMC9720743 DOI: 10.1242/dev.200683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 08/31/2022] [Indexed: 03/31/2024]
Abstract
The binding of 17β-oestradiol to oestrogen receptor alpha (ERα) plays a crucial role in the control of reproduction, acting through both nuclear and membrane-initiated signalling. To study the physiological role of membrane ERα in the reproductive system, we used the C451A-ERα mouse model with selective loss of function of membrane ERα. Despite C451A-ERα mice being described as sterile, daily weighing and ultrasound imaging revealed that homozygous females do become pregnant, allowing the investigation of the role of ERα during pregnancy for the first time. All neonatal deaths of the mutant offspring mice resulted from delayed parturition associated with failure in pre-term progesterone withdrawal. Moreover, pregnant C451A-ERα females exhibited partial intrauterine embryo arrest at about E9.5. The observed embryonic lethality resulted from altered expansion of Tpbpa-positive spiral artery-associated trophoblast giant cells into the utero-placental unit, which is associated with an imbalance in expression of angiogenic factors. Together, these processes control the trophoblast-mediated spiral arterial remodelling. Hence, loss of membrane ERα within maternal tissues clearly alters the activity of invasive trophoblast cells during placentogenesis. This previously unreported function of membrane ERα could open new avenues towards a better understanding of human pregnancy-associated pathologies.
Collapse
Affiliation(s)
- Mariam Rusidzé
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297-UPS, CHU, Toulouse 31432, France
| | | | - Pierre Sicard
- IPAM, BioCampus Montpellier, CNRS, INSERM, University of Montpellier, Montpellier 34295, France
| | - Isabelle Raymond-Letron
- Institut Restore, Université de Toulouse, CNRS U-5070, EFS, ENVT, Inserm U1031, Toulouse 31076, France
| | - Frank Giton
- APHP H.Mondor - IMRB - INSERM U955, Créteil 94010, France
| | - Emilie Vessieres
- Angers University, MITOVASC, CarMe team, CNRS UMR 6015, INSERM U1083, Angers 49055, France
| | - Vincent Prevot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille 59000, France
| | - Daniel Henrion
- Angers University, MITOVASC, CarMe team, CNRS UMR 6015, INSERM U1083, Angers 49055, France
| | | | | | - Françoise Lenfant
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297-UPS, CHU, Toulouse 31432, France
| |
Collapse
|
34
|
Trinh QD, Takada K, Pham NTK, Takano C, Namiki T, Ikuta R, Hayashida S, Okitsu S, Ushijima H, Komine-Aizawa S, Hayakawa S. Enhancement of Rubella Virus Infection in Immortalized Human First-Trimester Trophoblasts Under Low-Glucose Stress Conditions. Front Microbiol 2022; 13:904189. [PMID: 35875557 PMCID: PMC9304883 DOI: 10.3389/fmicb.2022.904189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/21/2022] [Indexed: 12/25/2022] Open
Abstract
Rubella virus (RuV) infections in pregnant women, especially first-trimester infections, can lead to congenital rubella syndrome (CRS). However, the mechanisms of fetal RuV infection are not completely understood, and it is not observed in every pregnant woman infected with RuV. As gestational diabetes mellitus is a risk factor for congenital viral infections, we investigated the possible roles of hypoglycemia-related endoplasmic reticulum (ER) stress as a key factor for vertical RuV infection using immortalized human first-trimester trophoblasts. Low-glucose stress was induced prior to RuV infection by culturing HTR-8/SVneo and Swan.71 cells in low-glucose (LG) medium for 24 h or high-glucose medium for 6 h and then LG medium for an additional 18 h. Clinically isolated RuV was inoculated at a multiplicity of infection of 5 to 10. The intracellular localization of the RuV capsid protein was investigated 24 to 48 h post-infection (pi) with flow cytometry (FCM) analysis and fluorescence microscopy. Viral progeny production was monitored by FCM analysis. Increases in RuV infection in LG-induced ER-stressed trophoblasts were observed. No significant increase in apoptosis of RuV-infected cells was noted at days 2 and 5 pi, and substantial viral progeny production was observed until day 5 pi. An approximate fivefold increase in viral binding was noted for the LG-stressed cells. Although the detailed mechanisms underlying viral entry into LG-stressed cells are not known and require further investigation, these findings suggest that a certain degree of LG stress in early pregnancy may facilitate infection and cause CRS.
Collapse
Affiliation(s)
- Quang Duy Trinh
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Ngan Thi Kim Pham
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Chika Takano
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | | | - Ryo Ikuta
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo, Japan
| | | | - Shoko Okitsu
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Hiroshi Ushijima
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Shihoko Komine-Aizawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- *Correspondence: Shihoko Komine-Aizawa,
| | - Satoshi Hayakawa
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Satoshi Hayakawa,
| |
Collapse
|
35
|
Monteiro VRS, Andrade CBV, Gomes HR, Reginatto MW, Império GE, Fontes KN, Spiess DA, Rangel-Junior WS, Nascimento VMO, Lima COS, Sousa RPC, Bloise FF, Matthews SG, Bloise E, Pimentel-Coelho PM, Ortiga-Carvalho TM. Mid-pregnancy poly(I:C) viral mimic disrupts placental ABC transporter expression and leads to long-term offspring motor and cognitive dysfunction. Sci Rep 2022; 12:10262. [PMID: 35715474 PMCID: PMC9205917 DOI: 10.1038/s41598-022-14248-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/03/2022] [Indexed: 11/09/2022] Open
Abstract
Limited information is available about the effect of mid-pregnancy viral infections on the placental expression of efflux transporters and offspring behavior. We hypothesized that maternal exposure to polyinosinic-polycytidylic acid [poly(I:C)], a synthetic double-stranded RNA viral mimic, would impair placental cell turnover, the expression of selected ABC transporters and adult offspring behavior. C57BL/6 mice were administered poly(I:C) (10 mg/Kg;ip) or vehicle at gestational day (GD) 13.5 (mid-pregnancy). Dams were euthanized for blood collection 4 h after injection, fetal and placental collection at GD18.5 or allowed to deliver spontaneously at term. At GD 13.5, poly(I:C) induced an acute pro-inflammatory response characterized by an increase in maternal plasma levels of IL-6, CXCL-1 and CCL-2/MCP-1. At GD 18.5, poly(I:C) decreased cell proliferation/death in the labyrinthine and increased cell death in the junctional zones, characterizing a disruption of placental cell turnover. Abca1 and Abcg1 immunolabelling was decreased in the labyrinthine zone, whereas Abca1, Abcg1 and breast cancer resistance transporter (Bcrp) expression increased in the junctional zone. Moreover, adult offspring showed motor and cognitive impairments in the Rotarod and T-water maze tests. These results indicate that viral infection during mid-pregnancy may disrupt relevant placental efflux transporters, as well as placental cell turnover and offspring behavior in adult life.
Collapse
Affiliation(s)
- V R S Monteiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - C B V Andrade
- Departamento de Histologia e Embriologia, Instituto de Biologia Roberto Alcantara Gomes, Universidade Estadual Do Rio de Janeiro, Rio de Janeiro, Brasil
| | - H R Gomes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - M W Reginatto
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - G E Império
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - K N Fontes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - D A Spiess
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - W S Rangel-Junior
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - V M O Nascimento
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - C O S Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - R P C Sousa
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - F F Bloise
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - S G Matthews
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Canada.,Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Toronto, Toronto, Canada.,Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, Canada
| | - E Bloise
- Departamento de Morfologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | - P M Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - T M Ortiga-Carvalho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil.
| |
Collapse
|
36
|
Abstract
Cardiovascular complications of pregnancy have risen substantially over the past decades, and now account for the majority of pregnancy-induced maternal deaths, as well as having substantial long-term consequences on maternal cardiovascular health. The causes and pathophysiology of these complications remain poorly understood, and therapeutic options are limited. Preclinical models represent a crucial tool for understanding human disease. We review here advances made in preclinical models of cardiovascular complications of pregnancy, including preeclampsia and peripartum cardiomyopathy, with a focus on pathological mechanisms elicited by the models and on relevance to human disease.
Collapse
Affiliation(s)
- Zolt Arany
- Department of Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia (Z.A.)
| | - Denise Hilfiker-Kleiner
- Institute of Cardiovascular Complications in Pregnancy and in Oncologic Therapies, Philipps University Marburg, Germany (D.H.-K.)
| | - S Ananth Karumanchi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (S.A.K.)
| |
Collapse
|
37
|
Eikmans M, van der Keur C, Anholts JDH, Drabbels JJM, van Beelen E, de Sousa Lopes SMC, van der Hoorn ML. Primary Trophoblast Cultures: Characterization of HLA Profiles and Immune Cell Interactions. Front Immunol 2022; 13:814019. [PMID: 35634345 PMCID: PMC9136060 DOI: 10.3389/fimmu.2022.814019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 04/11/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction Trophoblasts are essential in fetal-maternal interaction during pregnancy. The goal was to study HLA profiles of primary trophoblasts derived from placentas, and to investigate their usefulness in studying interaction with immune cells. Methods After enzymatic digestion of first-trimester placental tissue from seven donors (6-9 weeks gestation) and trophoblast enrichment we cultured cytotrophoblasts (CTB) in stem cell medium. CTB were differentiated into EVT in a Matrigel-containing medium. A subset of CTB/EVT was profiled for microRNA levels. Expression of classical HLA molecules and of HLA-G was studied by flow cytometry, qPCR, and ELISA. Secondary trophoblast cell lines JAR and JEG-3 were studied as controls. Lymphocytes were investigated during co-culturing with EVT. Results The trophoblasts could be easily maintained for several passages, upregulated classical trophoblast markers (GATA3, TFAP2C, chromosome-19 microRNAs), and upon differentiation to EVT they were selective in expressing HLA-C. EVT showed increasing expression of total HLA-G, an increasing proportion of HLA-G1 over G2- and G3 isoforms, and elevated excretion of soluble HLA-G. These features were distinct from those of the secondary trophoblast cell lines. TNF-α and IL-8 represented the most abundantly secreted cytokines by CTB, but their levels were minimal in EVT cultures. As proof of principle, we showed that EVT affect lymphocytes in three-day co-cultures (n=4) by decreasing activation marker HLA-DR. Conclusion We verified the possibility culturing trophoblasts from first-term placentas, and their capability of differentiating to HLA-G expressing EVT. This culture model better represents the in-vivo situation than previously studied secondary trophoblast cell lines and enables mechanistic studies of fetal-maternal interactions.
Collapse
Affiliation(s)
- Michael Eikmans
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- *Correspondence: Michael Eikmans,
| | - Carin van der Keur
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Jos J. M. Drabbels
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Els van Beelen
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | | | | |
Collapse
|
38
|
Small RNA sequencing reveals placenta-derived exosomal microRNAs associated with preeclampsia. J Hypertens 2022; 40:1030-1041. [PMID: 35202022 DOI: 10.1097/hjh.0000000000003112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Preeclampsia is one of the most important complications during pregnancy and the leading cause of maternal morbidity and mortality; however, the pathogenesis of preeclampsia remains partially misunderstood. The aim of this study was to identify placenta-derived exosomal microRNAs (miRNAs) involved in the preeclampsia process. METHODS Peripheral blood was collected from normal and preeclampsia pregnant women, and placenta-derived exosomes were extracted. Small RNA sequencing was performed to identify the exosomal miRNAs involved in preeclampsia. The function of a differentially expressed exosomal miRNA was verified. RESULTS The extracted exosomes presented round or oval-like structures with diameters of approximately 80 nm and could be recognized by antibodies against CD9, CD81, and placental alkaline phosphatase. A total of 1013 exosomal miRNAs were identified by small RNA sequencing, of which 946 were known miRNAs and 67 were novel miRNAs. Twenty-six miRNAs were identified as differentially expressed when comparing the data of the preeclampsia and normal groups. One of the differentially expressed miRNAs, hsa-miR-370-3p, which was upregulated in the preeclampsia group, was shown to bind to the 3' untranslated region of C-X-C motif chemokine 12, a chemokine that plays important role during preeclampsia process. Moreover, functional analysis revealed that hsa-miR-370-3p could inhibit proliferation, migration, and invasion while promoting apoptosis of HTR-8/SVneo cells. CONCLUSION A total of 1013 placenta-derived exosomal miRNAs were identified by small RNA sequencing, of which 26 were differentially expressed. The function of one differentially expressed miRNA (hsa-miR-370-3p) was verified. Our results provide new perspectives on the pathogenesis of preeclampsia and potential biomarkers for preeclampsia diagnosis.
Collapse
|
39
|
Santos BR, Dos Anjos Cordeiro JM, Santos LC, de Oliveira LS, Mendonça LD, Santos EO, de Macedo IO, Szawka RE, Serakides R, Silva JF. Maternal hypothyroidism reduces the expression of the kisspeptin/Kiss1r system in the maternal-fetal interface of rats. Reprod Biol 2022; 22:100615. [PMID: 35180577 DOI: 10.1016/j.repbio.2022.100615] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 01/13/2022] [Accepted: 02/05/2022] [Indexed: 11/16/2022]
Abstract
Alterations of circulating and placental levels of kisspeptin have been associated with gestational diseases. However, there are still no studies on the placental and decidual expression of Kiss1 and its receptor Kiss1r in maternal hypothyroidism, which is the aim of this work. We demonstrate that the fetoplacental restriction caused by hypothyroidism in rats is associated with a reduction in the Kiss1r expression and reduced Kiss1 and Kiss1r mRNA levels in the decidua and/or placenta. This demonstrate that fetoplacental restriction in hypothyroid rats is linked with a suppression of the kisspeptin/Kiss1r system at the maternal-fetal interface.
Collapse
Affiliation(s)
- Bianca Reis Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Jeane Martinha Dos Anjos Cordeiro
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Luciano Cardoso Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Luciana Santos de Oliveira
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Letícia Dias Mendonça
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Emilly Oliveira Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Isabella Oliveira de Macedo
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil
| | - Raphael Escorsim Szawka
- Departamento de Fisiologia e Biofísica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Rogéria Serakides
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, 31270-901, Belo Horizonte, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, 45662-900, Ilheus, Brazil.
| |
Collapse
|
40
|
Garner KL, Bowdridge EC, Griffith JA, DeVallance E, Seman MG, Engels KJ, Groth CP, Goldsmith WT, Wix K, Batchelor TP, Nurkiewicz TR. Maternal Nanomaterial Inhalation Exposure: Critical Gestational Period in the Uterine Microcirculation is Angiotensin II Dependent. Cardiovasc Toxicol 2022; 22:167-180. [PMID: 35066857 PMCID: PMC9013006 DOI: 10.1007/s12012-021-09712-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/07/2021] [Indexed: 11/03/2022]
Abstract
Maternal inhalation exposure to engineered nanomaterials (ENM) has been associated with microvascular dysfunction and adverse cardiovascular responses. Pregnancy requires coordinated vascular adaptation and growth that are imperative for survival. Key events in pregnancy hallmark distinct periods of gestation such as implantation, spiral artery remodeling, placentation, and trophoblast invasion. Angiotensin II (Ang II) is a critical vasoactive mediator responsible for adaptations and is implicated in the pathology of preeclampsia. If perturbations occur during gestation, such as those caused by ENM inhalation exposure, then maternal-fetal health consequences may occur. Our study aimed to identify the period of gestation in which maternal microvascular functional and fetal health are most vulnerable. Additionally, we wanted to determine if Ang II sensitivity and receptor density is altered due to exposure. Dams were exposed to ENM aerosols (nano-titanium dioxide) during three gestational windows: early (EE, gestational day (GD) 2-6), mid (ME, GD 8-12) or late (LE, GD 15-19). Within the EE group dry pup mass decreased by 16.3% and uterine radial artery wall to lumen ratio (WLR) increased by 25.9%. Uterine radial artery response to Ang II sensitivity increased by 40.5% in the EE group. Ang II receptor density was altered in the EE and LE group with decreased levels of AT2R. We conclude that early gestational maternal inhalation exposures resulted in altered vascular anatomy and physiology. Exposure during this time-period results in altered vascular reactivity and changes to uterine radial artery WLR, leading to decreased perfusion to the fetus and resulting in lower pup mass.
Collapse
Affiliation(s)
- Krista L Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth C Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Julie A Griffith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Evan DeVallance
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Madison G Seman
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Kevin J Engels
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Caroline P Groth
- Department of Epidemiology and Biostatistics, West Virginia University School of Public Health, Morgantown, WV, USA
| | - William T Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Kim Wix
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thomas P Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA
| | - Timothy R Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, Morgantown, WV, USA.
- Center for Inhalation Toxicology (iTOX), West Virginia University School of Medicine, Morgantown, WV, USA.
- Department of Physiology and Pharmacology, Robert C. Byrd Health Sciences Center, West Virginia University, 64 Medical Center Drive, Morgantown, WV, 26506-9229, USA.
| |
Collapse
|
41
|
ietary curcumin supplementation ameliorates placental inflammation in rats with intra-uterine growth retardation by inhibiting the NF-κB signaling pathway. J Nutr Biochem 2022; 104:108973. [DOI: 10.1016/j.jnutbio.2022.108973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/18/2021] [Accepted: 01/31/2022] [Indexed: 12/21/2022]
|
42
|
Expression of tissue factor and TF-mediated integrin regulation in HTR-8/SVneo trophoblast cells. J Reprod Immunol 2022; 150:103473. [PMID: 35030354 DOI: 10.1016/j.jri.2022.103473] [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: 07/23/2021] [Revised: 10/16/2021] [Accepted: 01/03/2022] [Indexed: 11/22/2022]
Abstract
Placenta is a crucial source of Tissue Factor (TF) to initiate coagulation. As far as the TF is concern, aberrant expression of TF has been reported to have a significant role in thrombosis, inflammation, cancer metastasis and atherosclerosis. It is evident that TF and TF-FVIIa complex has major roles in the disease process beyond hemostasis and thrombosis. On the other hand, TF-FVII-dependent signaling primarily activates PAR2 and inducing pro-angiogenic and immune-modulating cytokines in tumor environment. However, the role of TF has not been delineated in placental functions. Integrin typically binds to the extracellular matrix which in turn mediate cell-cell adhesion and cell behavior for migration. Dysregulation of integrin expression affects cell interaction, proliferation, and migration. Therefore, this study aims to ascertain the expression of TF in HTR-8/SVneo trophoblast cell line and its role in signal transduction of integrin (ITGα1, ITGα2, ITGβ1) regulation concerning the invasion of trophoblasts. We have used RT-PCR and Western blot for the gene and protein expression analysis respectively. In addition, cell migration assays, MTT, and DAPI were performed to examine migration, cytotoxicity and apoptosis effect of FVIIa. The results suggest that the gene and protein level expressions of TF were predominant in HTR-8/SVneo cell line. Further, the cytotoxicity and apoptosis in HTR-8/SVneo cells were not observed when treated with FVIIa. The cells treated with FVIIa shown a dose-dependent up-regulation of integrin(s) (**p < 0.01, *p < 0.05) when compared to control. Migration of the HTR-8/SVneo cells was observed without any apoptosis in FVIIa-treated cells when compared to that of control. On the whole, these observations delineated the TF-FVIIa interaction in modulating the TF-dependent integrin signal transduction in HTR-8/SVneo trophoblast cell line.
Collapse
|
43
|
Martín-Estal I, Castorena-Torres F. Gestational Diabetes Mellitus and Energy-Dense Diet: What Is the Role of the Insulin/IGF Axis? Front Endocrinol (Lausanne) 2022; 13:916042. [PMID: 35813659 PMCID: PMC9259869 DOI: 10.3389/fendo.2022.916042] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/24/2022] [Indexed: 01/02/2023] Open
Abstract
Gestational diabetes mellitus (GDM), is one of the most important pregnancy complications affecting approximately 15% of pregnant women. It is related to several gestational adverse outcomes in the fetus, e.g., macrosomia, shoulder dystocia, stillbirth, neonatal hypoglycemia, and respiratory distress. Women with GDM have a high risk of developing type 2 diabetes in the future. The pathogenesis of GDM is not completely understood; nevertheless, two factors could contribute to its development: β-cell dysfunction and failure in insulin secretion in response to insulin resistance induced by gestation. Both processes, together with the physiological activities of the insulin-like growth factors (IGFs), play a crucial role in glucose transport to the fetus and hence, fetal growth and development. IGFs (both IGF-1 and IGF-2) and their binding proteins (IGFBPs) regulate glucose metabolism and insulin sensitivity. Maternal nutritional status determines the health of the newborn, as it has substantial effects on fetal growth and development. Maternal obesity and an energy-dense diet can cause an increase in insulin and IGF-1 serum levels, producing metabolic disorders, such as insulin resistance, GDM, and high birth weight (> 4,000 g) due to a higher level of body fat. In this way, in GDM pregnancies there is an increase in IGF-1 and IGF-2 serum levels, and a decrease in IGFBP-1 and 4 serum levels, suggesting the crucial role of the insulin/IGF system in this gestational outcome. Here, the present review tries to elucidate the role that energy-dense diets and the insulin/IGF-1 signaling pathway perform in GDM pregnancies.
Collapse
|
44
|
Matić M, Obradović A, Milošević M, Paunović M, Ognjanović B. The effects of Interleukin-6 on viability, redox homeostasis and migration capacity of human placental cells JEG-3 in chemically induced hypoxia. KRAGUJEVAC JOURNAL OF SCIENCE 2022. [DOI: 10.5937/kgjsci2244103m] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Interleukin-6 (IL-6) is a pleiotropic cytokine involved in the regulation of cell growth and differentiation exerting an important role in the immune and inflammatory response. This study aimed to evaluate the effects of three increasing concentrations of IL-6 (1 pg/mL, 5 pg/mL, and 10 pg/mL) on cell viability, redox homeostasis parameters (O2∙, NO2-, glutathione) and migratory potential in human trophoblast cell line JEG-3 under chemically induced hypoxia in short-term (24 h) and long-term (72 h) exposure. The obtained results show a dose-dependent reduction of cell viability and NO levels, while the concentration of O2 ∙increased. Levels of total glutathione increased in a dose-dependent manner compared to control cells, suggesting its significant antioxidative contribution in hypoxic conditions. The migratory potential of cells was significantly elevated in the two highest applied doses implying the disturbance of cell invasive homeostasis at its pathological concentrations, which could represent a risk factor in some pregnancy disorders.
Collapse
|
45
|
Santos BR, dos Anjos Cordeiro JM, Santos LC, Barbosa EM, Mendonça LD, Santos EO, de Macedo IO, de Lavor MSL, Szawka RE, Serakides R, Silva JF. Kisspeptin treatment improves fetal-placental development and blocks placental oxidative damage caused by maternal hypothyroidism in an experimental rat model. Front Endocrinol (Lausanne) 2022; 13:908240. [PMID: 35966095 PMCID: PMC9365946 DOI: 10.3389/fendo.2022.908240] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/04/2022] [Indexed: 12/02/2022] Open
Abstract
Maternal hypothyroidism is associated with fetal growth restriction, placental dysfunction, and reduced kisspeptin/Kiss1R at the maternal-fetal interface. Kisspeptin affects trophoblastic migration and has antioxidant and immunomodulatory activities. This study aimed to evaluate the therapeutic potential of kisspeptin in the fetal-placental dysfunction of hypothyroid Wistar rats. Hypothyroidism was induced by daily administration of propylthiouracil. Kisspeptin-10 (Kp-10) treatment was performed every other day or daily beginning on day 8 of gestation. Feto-placental development, placental histomorphometry, and expression levels of growth factors (VEGF, PLGF, IGF1, IGF2, and GLUT1), hormonal (Dio2) and inflammatory mediators (TNFα, IL10, and IL6), markers of hypoxia (HIF1α) and oxidative damage (8-OHdG), antioxidant enzymes (SOD1, Cat, and GPx1), and endoplasmic reticulum stress mediators (ATF4, GRP78, and CHOP) were evaluated on day 18 of gestation. Daily treatment with Kp-10 increased free T3 and T4 levels and improved fetal weight. Both treatments reestablished the glycogen cell population in the junctional zone. Daily treatment with Kp-10 increased the gene expression levels of Plgf, Igf1, and Glut1 in the placenta of hypothyroid animals, in addition to blocking the increase in 8-OHdG and increasing protein and/or mRNA expression levels of SOD1, Cat, and GPx1. Daily treatment with Kp-10 did not alter the higher protein expression levels of VEGF, HIF1α, IL10, GRP78, and CHOP caused by hypothyroidism in the junctional zone compared to control, nor the lower expression of Dio2 caused by hypothyroidism. However, in the labyrinth zone, this treatment restored the expression of VEGF and IL10 and reduced the GRP78 and CHOP immunostaining. These findings demonstrate that daily treatment with Kp-10 improves fetal development and placental morphology in hypothyroid rats, blocks placental oxidative damage, and increases the expression of growth factors and antioxidant enzymes in the placenta.
Collapse
Affiliation(s)
- Bianca Reis Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
| | - Jeane Martinha dos Anjos Cordeiro
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
| | - Luciano Cardoso Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
| | - Erikles Macedo Barbosa
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
| | - Letícia Dias Mendonça
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
| | - Emilly Oliveira Santos
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
| | - Isabella Oliveira de Macedo
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
| | - Mário Sergio Lima de Lavor
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
| | - Raphael Escorsim Szawka
- Departamento de Fisiologia e Biofísica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rogeria Serakides
- Departamento de Clinica e Cirurgia Veterinarias, Escola de Veterinaria, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletronica, Departamento de Ciencias Biologicas, Universidade Estadual de Santa Cruz, Campus Soane Nazare de Andrade, Ilheus, Brazil
- *Correspondence: Juneo Freitas Silva,
| |
Collapse
|
46
|
Meakin C, Barrett ES, Aleksunes LM. Extravillous trophoblast migration and invasion: Impact of environmental chemicals and pharmaceuticals. Reprod Toxicol 2022; 107:60-68. [PMID: 34838982 PMCID: PMC8760155 DOI: 10.1016/j.reprotox.2021.11.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/09/2021] [Accepted: 11/22/2021] [Indexed: 01/03/2023]
Abstract
During pregnancy, the migration and invasion of extravillous trophoblasts (EVTs) into the maternal uterus is essential for proper development of the placenta and fetus. During the first trimester, EVTs engraft and remodel maternal spiral arteries allowing for efficient blood flow and the transfer of essential nutrients and oxygen to the fetus. Aberrant migration of EVTs leading to either shallow or deep invasion into the uterus has been implicated in a number of gestational pathologies including preeclampsia, fetal growth restriction, and placenta accreta spectrum. The migration and invasion of EVTs is well-coordinated to ensure proper placentation. However, recent data point to the ability of xenobiotics to disrupt EVT migration. These xenobiotics include heavy metals, endocrine disrupting chemicals, and organic contaminants and have often been associated with adverse pregnancy outcomes. In most instances, xenobiotics appear to reduce EVT migration; however, there are select examples of enhanced motility after chemical exposure. In this review, we provide an overview of the 1) current experimental approaches used to evaluate EVT migration and invasion in vitro, 2) ability of environmental chemicals and pharmaceuticals to enhance or retard EVT motility, and 3) signaling pathways responsible for altered EVT migration that are sensitive to disruption by xenobiotics.
Collapse
Affiliation(s)
- Cassandra Meakin
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ
| | - Emily S. Barrett
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ,Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ
| | - Lauren M. Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ,Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ,Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ,Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ
| |
Collapse
|
47
|
Ali A, Alexander S, Ko P, Cuffe JSM, Whitehouse AJO, McGrath JJ, Eyles D. Developmental Vitamin D Deficiency in Pregnant Rats Does Not Induce Preeclampsia. Nutrients 2021; 13:4254. [PMID: 34959804 PMCID: PMC8707812 DOI: 10.3390/nu13124254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022] Open
Abstract
Preeclampsia is a pregnancy disorder characterized by hypertension. Epidemiological studies have associated preeclampsia with an increased risk of neurodevelopmental disorders in offspring, such as autism and schizophrenia. Preeclampsia has also been linked with maternal vitamin D deficiency, another candidate risk factor also associated with autism. Our laboratory has established a gestational vitamin-D-deficient rat model that shows consistent and robust behavioural phenotypes associated with autism- and schizophrenia-related animal models. Therefore, we explored here whether this model also produces preeclampsia as a possible mediator of behavioural phenotypes in offspring. We showed that gestational vitamin D deficiency was not associated with maternal blood pressure or proteinuria during late gestation. Maternal and placental angiogenic and vasculogenic factors were also not affected by a vitamin-D-deficient diet. We further showed that exposure to low vitamin D levels did not expose the placenta to oxidative stress. Overall, gestational vitamin D deficiency in our rat model was not associated with preeclampsia-related features, suggesting that well-described behavioural phenotypes in offspring born to vitamin-D-deficient rat dams are unlikely to be mediated via a preeclampsia-related mechanism.
Collapse
Affiliation(s)
- Asad Ali
- Neurobiology, Queensland Brain Institute, University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (S.A.); (P.K.); (J.J.M.)
| | - Suzanne Alexander
- Neurobiology, Queensland Brain Institute, University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (S.A.); (P.K.); (J.J.M.)
- Neurobiology, Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia
| | - Pauline Ko
- Neurobiology, Queensland Brain Institute, University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (S.A.); (P.K.); (J.J.M.)
- Neurobiology, Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia
| | - James S. M. Cuffe
- Placental Endocrinology, School of Biomedical Sciences, University of Queensland, St. Lucia, QLD 4072, Australia;
| | - Andrew J. O. Whitehouse
- Autism Research Team, Telethon Kids Institute, The University of Western Australia, Crawley, WA 6009, Australia;
| | - John J. McGrath
- Neurobiology, Queensland Brain Institute, University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (S.A.); (P.K.); (J.J.M.)
- Neurobiology, Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia
- NCRR—National Centre for Register-Based Research, Department of Economics and Business Economics, Aarhus University, 8000 Aarhus, Denmark
| | - Darryl Eyles
- Neurobiology, Queensland Brain Institute, University of Queensland, St. Lucia, QLD 4072, Australia; (A.A.); (S.A.); (P.K.); (J.J.M.)
- Neurobiology, Queensland Centre for Mental Health Research, Wacol, QLD 4076, Australia
| |
Collapse
|
48
|
Fuchs J, Nonn O, Daxboeck C, Groiss S, Moser G, Gauster M, Lang-Olip I, Brislinger D. Automated Quantitative Image Evaluation of Antigen Retrieval Methods for 17 Antibodies in Placentation and Implantation Diagnostic and Research. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2021; 27:1-12. [PMID: 34851247 DOI: 10.1017/s1431927621012630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Immunostaining in clinical routine and research highly depends on standardized staining methods and quantitative image analyses. We qualitatively and quantitatively compared antigen retrieval methods (no pretreatment, pretreatment with pepsin, and heat-induced pretreatment with pH 6 or pH 9) for 17 antibodies relevant for placenta and implantation diagnostics and research. Using our newly established, comprehensive automated quantitative image analysis approach, fluorescent signal intensities were evaluated. Automated quantitative image analysis found that 9 out of 17 antibodies needed antigen retrieval to show positive staining. Heat induction proved to be the most efficient form of antigen retrieval. Eight markers stained positive after pepsin digestion, with β-hCG and vWF showing enhanced staining intensities. To avoid the misinterpretation of quantitative image data, the qualitative aspect should always be considered. Results from native placental tissue were compared with sections of a placental invasion model based on thermo-sensitive scaffolds. Immunostaining on placentas in vitro leads to new insights into fetal development and maternal pathophysiological pathways, as pregnant women are justifiably excluded from clinical studies. Thus, there is a clear need for the assessment of reliable immunofluorescent staining and pretreatment methods. Our evaluation offers a powerful tool for antibody and pretreatment selection in placental research providing objective and precise results.
Collapse
Affiliation(s)
- Julia Fuchs
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, GrazA-8010, Austria
| | - Olivia Nonn
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, GrazA-8010, Austria
| | - Christine Daxboeck
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, GrazA-8010, Austria
| | - Silvia Groiss
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, GrazA-8010, Austria
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, GrazA-8010, Austria
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, GrazA-8010, Austria
| | - Ingrid Lang-Olip
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, GrazA-8010, Austria
| | - Dagmar Brislinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, GrazA-8010, Austria
| |
Collapse
|
49
|
Li A, Li S, Zhang C, Fang Z, Sun Y, Peng Y, Wang X, Zhang M. FPR2 serves a role in recurrent spontaneous abortion by regulating trophoblast function via the PI3K/AKT signaling pathway. Mol Med Rep 2021; 24:838. [PMID: 34608500 PMCID: PMC8503740 DOI: 10.3892/mmr.2021.12478] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/10/2021] [Indexed: 01/02/2023] Open
Abstract
Recurrent spontaneous abortion (RSA) effects both the physical and mental health of women of reproductive age. Trophoblast dysfunction may result in RSA due to shallow placental implantation. The mechanisms underlying formyl peptide receptor 2 (FPR2) on the biological functions of trophoblasts remain to be elucidated. The present study aimed to explore the potential functions of FPR2, a G protein‑coupled receptor, in placental trophoblasts. The location and expression levels of FPR2 in the villi tissue of patients with RSA were detected using immunohistochemical staining, reverse transcription‑quantitative PCR and western blotting. Following the transfection of small interfering RNA targeting FPR2 in HTR‑8/SVneo cells, a Cell Counting Kit‑8 assay was used to determine the levels of cell viability. Flow cytometry was used to examine the levels of cell apoptosis and gap closure and Transwell assays were carried out to evaluate the levels of cell migration and invasion. A tube formation assay was performed to detect the levels of capillary‑like structure formation. Western blotting was used to detect the expression levels of proteins in the associated signaling pathways. The expression of FPR2 was present in villi trophoblasts and was markedly increased in patients with RSA. The levels of trophoblast invasion, migration and tube formation were markedly increased following FPR2 knockdown, whereas the levels of apoptosis were markedly decreased. In addition, FPR2 knockdown caused an increase in the phosphorylation levels of AKT and PI3K. Thus, FPR2 may be involved in the regulation of trophoblast function via the PI3K/AKT signaling pathway. The results of the present study provided a theoretical basis for the use of FPR2 as a target for the treatment of trophoblast‑associated diseases, such as RSA.
Collapse
Affiliation(s)
- Anna Li
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, Shandong 250014, P.R. China
| | - Shuxian Li
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, Shandong 250014, P.R. China
| | - Chongyu Zhang
- Department of Chronic Disease, Center for Disease Control and Prevention of Wulian, Rizhao, Shandong 262300, P.R. China
| | - Zhenya Fang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, Shandong 250014, P.R. China
| | - Yaqiong Sun
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, Shandong 250014, P.R. China
| | - Yanjie Peng
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, Shandong 250014, P.R. China
| | - Xietong Wang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, Shandong 250014, P.R. China
| | - Meihua Zhang
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
50
|
Kanter JR, Mani S, Gordon SM, Mainigi M. Uterine natural killer cell biology and role in early pregnancy establishment and outcomes. F&S REVIEWS 2021; 2:265-286. [PMID: 35756138 PMCID: PMC9232176 DOI: 10.1016/j.xfnr.2021.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Objective While immune cells were originally thought to only play a role in maternal tolerance of the semiallogenic fetus, an active role in pregnancy establishment is becoming increasingly apparent. Uterine natural killer (uNK) cells are of specific interest because of their cyclic increase in number during the window of implantation. As a distinct entity from their peripheral blood counterparts, understanding the biology and function of uNK cells will provide the framework for understanding their role in early pregnancy establishment and adverse pregnancy outcomes. Evidence Review This review discusses unique uNK cell characteristics and presents clinical implications resulting from their dysfunction. We also systematically present existing knowledge about uNK cell function in three processes critical for successful human embryo implantation and placentation: stromal cell decidualization, spiral artery remodeling, and extravillous trophoblast invasion. Finally, we review the features of uNK cells that could help guide future investigations. Results It is clear the uNK cells are intimately involved in multiple facets of early pregnancy. This is accomplished directly, through the secretion of factors that regulate stromal cells and trophoblast function; and indirectly, via interaction with other maternal cell types present at the maternal-fetal interface. Current work also suggests that uNK cells are a heterogenous population, with subsets that potentially accomplish different functions. Conclusion Establishment of pregnancy through successful embryo implantation and placentation requires crosstalk between multiple maternal cell types and invading fetal trophoblast cells. Defects in this process have been associated with multiple adverse perinatal outcomes including hypertensive disorders of pregnancy, placenta accreta, and recurrent miscarriage though the mechanism underlying development of these defects remain unclear. Abnormalities in NK cell number and function which would disrupt physiological maternal-fetal crosstalk, could play a critical role in abnormal implantation and placentation. It is therefore imperative to dissect the unique physiological role of uNK cells in pregnancy and use this knowledge to inform clinical practice by determining how uNK cell dysfunction could lead to reproductive failure.
Collapse
Affiliation(s)
- Jessica R. Kanter
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sneha Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Scott M. Gordon
- Division of Neonatology, Children’s Hospital of Philadelphia, Pennsylvania
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|