1
|
Jacobs A, Al-Juboori SI, Dobrinskikh E, Bolt MA, Sammel MD, Lijewski V, Post MD, Small JM, Su EJ. Placental differences between severe fetal growth restriction and hypertensive disorders of pregnancy requiring early preterm delivery: morphometric analysis of the villous tree supported by artificial intelligence. Am J Obstet Gynecol 2024; 231:552.e1-552.e13. [PMID: 38423447 PMCID: PMC11347726 DOI: 10.1016/j.ajog.2024.02.291] [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: 08/07/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND The great obstetrical syndromes of fetal growth restriction and hypertensive disorders of pregnancy can occur individually or be interrelated. Placental pathologic findings often overlap between these conditions, regardless of whether 1 or both diagnoses are present. Quantification of placental villous structures in each of these settings may identify distinct differences in developmental pathways. OBJECTIVE This study aimed to determine how the quantity and surface area of placental villi and vessels differ between severe, early-onset fetal growth restriction with absent or reversed umbilical artery Doppler indices and hypertensive disorders of pregnancy or the 2 conditions combined among subjects with disease severity that warrant early preterm delivery. We hypothesized that the trajectories of placental morphogenesis diverge after a common initiating insult of deep defective placentation. Specifically, we postulated that only villi are affected in pregnancy-related hypertension, whereas both villous and vascular structures are proportionally diminished in severe fetal growth restriction with no additional effect when hypertension is concomitantly present. STUDY DESIGN In this retrospective cohort study, paraffin-embedded placental tissue was obtained from 4 groups, namely (1) patients with severe fetal growth restriction with absent or reversed umbilical artery end-diastolic velocities and hypertensive disorders of pregnancy, (2) patients with severe fetal growth restriction with absent or reversed umbilical artery Doppler indices and no hypertension, (3) gestational age-matched, appropriately grown pregnancies with hypertensive disease, and (4) gestational age-matched, appropriately grown pregnancies without hypertension. Dual immunohistochemistry for cytokeratin-7 (trophoblast) and CD34 (endothelial cells) was performed, followed by artificial intelligence-driven morphometric analyses. The number of villi, total villous area, number of fetoplacental vessels, and total vascular area across villi within a uniform region of interest were quantified. Quantitative analyses of placental structures were modeled using linear regression. RESULTS Placentas from pregnancies complicated by hypertensive disorders of pregnancy exhibited significantly fewer stem villi (-282 stem villi; 95% confidence interval, -467 to -98; P<.01), a smaller stem villous area (-4.3 mm2; 95% confidence interval, -7.3 to -1.2; P<.01), and fewer stem villous vessels (-4967 stem villous vessels; 95% confidence interval, -8501 to -1433; P<.01) with no difference in the total vascular area. In contrast, placental abnormalities in cases with severe growth restriction were limited to terminal villi with global decreases in the number of villi (-873 terminal villi; 95% confidence interval, -1501 to -246; P<.01), the villous area (-1.5 mm2; 95% confidence interval, -2.7 to -0.4; P<.01), the number of blood vessels (-5165 terminal villous vessels; 95% confidence interval, -8201 to -2128; P<.01), and the vascular area (-0.6 mm2; 95% confidence interval, -1.1 to -0.1; P=.02). The combination of hypertension and growth restriction had no additional effect beyond the individual impact of each state. CONCLUSION Pregnancies complicated by hypertensive disorders of pregnancy exhibited defects in the stem villi only, whereas placental abnormalities in severely growth restricted pregnancies with absent or reversed umbilical artery end-diastolic velocities were limited to the terminal villi. There were no significant statistical interactions in the combination of growth restriction and hypertension, suggesting that distinct pathophysiological pathways downstream of the initial insult of defective placentation are involved in each entity and do not synergize to lead to more severe pathologic consequences. Delineating mechanisms that underly the divergence in placental development after a common inciting event of defective deep placentation may shed light on new targets for prevention or treatment.
Collapse
Affiliation(s)
- Anna Jacobs
- Rocky Vista University College of Osteopathic Medicine, Parker, CO
| | - Saif I Al-Juboori
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO
| | - Evgenia Dobrinskikh
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Matthew A Bolt
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Mary D Sammel
- Center for Innovative Design and Analysis, Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Virginia Lijewski
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO
| | - Miriam D Post
- Department of Pathology, University of Colorado School of Medicine, Aurora, CO
| | - James M Small
- Department of Biomedical Sciences; Rocky Vista University College of Osteopathic Medicine, Parker, CO
| | - Emily J Su
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO; Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO.
| |
Collapse
|
2
|
Shi JW, Lai ZZ, Zhou WJ, Yang HL, Zhang T, Sun JS, Zhao JY, Li MQ. TNFSF14 + natural killer cells prevent spontaneous abortion by restricting leucine-mediated decidual stromal cell senescence. EMBO J 2024; 43:5018-5036. [PMID: 39261664 PMCID: PMC11535022 DOI: 10.1038/s44318-024-00220-3] [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: 01/04/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 09/13/2024] Open
Abstract
In preparation for a potential pregnancy, the endometrium of the uterus changes into a temporary structure called the decidua. Senescent decidual stromal cells (DSCs) are enriched in the decidua during decidualization, but the underlying mechanisms of this process remain unclear. Here, we performed single-cell RNA transcriptomics on ESCs and DSCs and found that cell senescence during decidualization is accompanied by increased levels of the branched-chain amino acid (BCAA) transporter SLC3A2. Depletion of leucine, one of the branched-chain amino acids, from cultured media decreased senescence, while high leucine diet resulted in increased senescence and high rates of embryo loss in mice. BCAAs induced senescence in DSCs via the p38 MAPK pathway. In contrast, TNFSF14+ decidual natural killer (dNK) cells were found to inhibit DSC senescence by interacting with its ligand TNFRSF14. As in mice fed high-leucine diets, both mice with NK cell depletion and Tnfrsf14-deficient mice with excessive uterine senescence experienced adverse pregnancy outcomes. Further, we found excessive uterine senescence, SLC3A2-mediated BCAA intake, and insufficient TNFRSF14 expression in the decidua of patients with recurrent spontaneous abortion. In summary, this study suggests that dNK cells maintain senescence homeostasis of DSCs via TNFSF14/TNFRSF14, providing a potential therapeutic strategy to prevent DSC senescence-associated spontaneous abortion.
Collapse
Affiliation(s)
- Jia-Wei Shi
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, People's Republic of China
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China
- Department of Obstetrics and Gynecology, The first affiliated Hospital of Ningbo University, Ningbo, 315021, People's Republic of China
| | - Zhen-Zhen Lai
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China
| | - Wen-Jie Zhou
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Hui-Li Yang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China
| | - Tao Zhang
- Assisted Reproductive Technology Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Jian-Song Sun
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, 214122, People's Republic of China
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, People's Republic of China.
| | - Ming-Qing Li
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, People's Republic of China.
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, 200080, People's Republic of China.
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, People's Republic of China.
| |
Collapse
|
3
|
Zhong Y, Wang N, Lu S, Lu Y, Pan X, Zhou Y. Doppler Evaluation of Uterine Blood Flow in Patients with Unexplained Recurrent Pregnancy Loss. Int J Womens Health 2024; 16:1803-1814. [PMID: 39493660 PMCID: PMC11531733 DOI: 10.2147/ijwh.s477828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/12/2024] [Indexed: 11/05/2024] Open
Abstract
Objective This study aimed to analyze uterine artery and spiral artery hemodynamics in patients with unexplained recurrent pregnancy loss (URPL) with varying pregnancy outcomes. Methods 174 pregnant women with URPL and 144 pregnant women without adverse pregnancy histories were enrolled in this retrospective study. Based on pregnancy outcomes, these patients were divided into two groups: normal pregnancy outcomes (URPL-N, n=138) and adverse pregnancy outcomes (URPL-A, n=36). Control group participants were categorized into normal pregnancy outcomes (CON-N, n=129) and adverse pregnancy outcomes (CON-A, n=15). We compared uterine artery and spiral artery hemodynamics during different stages of gestation and the predictive value of these parameters for pregnancy outcomes. Results URPL-N group had fewer pregnancy losses and lower BMI compared to URPL-A group (P< 0.05). Spiral artery hemodynamics in URPL-N and CON-N groups were lower than those in URPL-A and CON-A groups during the mid-luteal phase, 11-13 weeks, 15-17 weeks, and 19-21 weeks of gestation, respectively. Uterine artery hemodynamics ((Pulsatility index (mPI), resistive index (mRI), and systolic-to-diastolic ratio (mS/D)) in the mid-luteal period were lower in URPL-N group than URPL-A group. Similarly, in CON-N group were lower than CON-A group. The URPL-A and CON-A groups had higher uterine artery and spiral artery hemodynamics when compared to the URPL-N and CON-N groups. Spiral artery hemodynamics exhibited larger areas under the ROC curve compared to uterine artery parameters. Conclusion Abnormal hemodynamics in these arteries may contribute to URPL and adverse pregnancy outcomes. Spiral artery hemodynamics are more reliable predictors of pregnancy outcomes than uterine artery parameters.
Collapse
Affiliation(s)
- Yanyu Zhong
- Reproductive Medicine Centre, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Nan Wang
- Reproductive Medicine Centre, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Sihui Lu
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Yaqian Lu
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Xin Pan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| | - Ying Zhou
- Reproductive Medicine Centre, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
4
|
Shukla V, Moreno-Irusta A, Varberg KM, Kuna M, Iqbal K, Galligos AM, Aplin JD, Choudhury RH, Okae H, Arima T, Soares MJ. NOTUM-mediated WNT silencing drives extravillous trophoblast cell lineage development. Proc Natl Acad Sci U S A 2024; 121:e2403003121. [PMID: 39325428 PMCID: PMC11459147 DOI: 10.1073/pnas.2403003121] [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: 02/14/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Trophoblast stem (TS) cells have the unique capacity to differentiate into specialized cell types, including extravillous trophoblast (EVT) cells. EVT cells invade into and transform the uterus where they act to remodel the vasculature facilitating the redirection of maternal nutrients to the developing fetus. Disruptions in EVT cell development and function are at the core of pregnancy-related disease. WNT-activated signal transduction is a conserved regulator of morphogenesis of many organ systems, including the placenta. In human TS cells, activation of canonical WNT signaling is critical for maintenance of the TS cell stem state and its downregulation accompanies EVT cell differentiation. We show that aberrant WNT signaling undermines EVT cell differentiation. Notum, palmitoleoyl-protein carboxylesterase (NOTUM), a negative regulator of canonical WNT signaling, was prominently expressed in first-trimester EVT cells developing in situ and up-regulated in EVT cells derived from human TS cells. Furthermore, NOTUM was required for optimal human TS cell differentiation to EVT cells. Activation of NOTUM in EVT cells is driven, at least in part, by endothelial Per-Arnt-Sim (PAS) domain 1 (also called hypoxia-inducible factor 2 alpha). Collectively, our findings indicate that canonical Wingless-related integration site (WNT) signaling is essential for maintenance of human trophoblast cell stemness and regulation of human TS cell differentiation. Downregulation of canonical WNT signaling via the actions of NOTUM is required for optimal EVT cell differentiation.
Collapse
Affiliation(s)
- Vinay Shukla
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Ayelen Moreno-Irusta
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Kaela M. Varberg
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Marija Kuna
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Khursheed Iqbal
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - Anna M. Galligos
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
| | - John D. Aplin
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, The University of Manchester, ManchesterM13 9WL, United Kingdom
- Manchester Academic Health Sciences Centre, St. Mary’s Hospital, University of Manchester, ManchesterM13 9WL, United Kingdom
| | - Ruhul H. Choudhury
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, The University of Manchester, ManchesterM13 9WL, United Kingdom
- Manchester Academic Health Sciences Centre, St. Mary’s Hospital, University of Manchester, ManchesterM13 9WL, United Kingdom
| | - Hiroaki Okae
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto860-0811, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai980-8575, Japan
| | - Michael J. Soares
- Department of Pathology and Laboratory Medicine, Institute for Reproductive and Developmental Sciences, University of Kansas Medical Center, Kansas City, KS66160
- Center for Perinatal Research, Children’s Research Institute, Children’s Mercy, Kansas City, MO64108
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS66160
| |
Collapse
|
5
|
Luo YH, Zhang YY, Li MQ, Zhang XY, Zheng ZM. Emerging Roles of IL-27 in Trophoblast Cells and Pregnancy Complications. Am J Reprod Immunol 2024; 92:e13942. [PMID: 39422056 DOI: 10.1111/aji.13942] [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: 05/06/2024] [Revised: 09/03/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
PROBLEM Pregnancy complications such as spontaneous abortion, preeclampsia, and preterm birth persist, despite current interventions aimed at their prevention and treatment largely proving unsuccessful. Interleukin-27 (IL-27), composed of p28 and EBI3 subunits, binds to IL-27R, which consists of gp130 and IL-27Rα (also known as WSX-1 or TCCR), and plays a pivotal role in tumor development and inflammation regulation. At the maternal-fetal interface, IL-27 expression has been detected in trophoblasts, endometrial stromal cells, and decidual cells. Abnormal levels of IL-27/IL-27R have been linked to adverse pregnancy outcomes, including spontaneous miscarriage, preeclampsia, and preterm birth. This review aims to explore the expression of IL-27 at the maternal-fetal interface and its signaling pathway, uncovering the complex role of IL-27 in pregnancy complications. METHOD OF STUDY A comprehensive literature review was conducted using PubMed/Medline, Scopus, and Embase databases, analyzing studies on IL-27 expression and its signaling pathways at the maternal-fetal interface. The review focused on identifying the presence of IL-27 in various cell types and linking abnormal IL-27/IL-27R expression to pregnancy complications such as spontaneous miscarriage, preeclampsia, and preterm birth. DISCUSSION AND CONCLUSION IL-27 plays a complex role at the maternal-fetal interface, with abnormal expression linked to several pregnancy complications. These findings highlight the need for further research to elucidate IL-27's mechanisms and develop targeted interventions. Future studies should aim to develop targeted interventions and improve therapeutic strategies for managing pregnancy complications.
Collapse
Affiliation(s)
- Yi-Hua Luo
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | - Yang-Yang Zhang
- Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, People's Republic of China
| | - Ming-Qing Li
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, Shanghai, People's Republic of China
| | - Xin-Yan Zhang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai, People's Republic of China
| | - Zi-Meng Zheng
- Department of Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Embryo Original Diseases, The International Peace Maternity and Child Health Hospital, Shanghai, People's Republic of China
| |
Collapse
|
6
|
Geisler HC, Safford HC, Mitchell MJ. Rational Design of Nanomedicine for Placental Disorders: Birthing a New Era in Women's Reproductive Health. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300852. [PMID: 37191231 PMCID: PMC10651803 DOI: 10.1002/smll.202300852] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/16/2023] [Indexed: 05/17/2023]
Abstract
The placenta is a transient organ that forms during pregnancy and acts as a biological barrier, mediating exchange between maternal and fetal circulation. Placental disorders, such as preeclampsia, fetal growth restriction, placenta accreta spectrum, and gestational trophoblastic disease, originate in dysfunctional placental development during pregnancy and can lead to severe complications for both the mother and fetus. Unfortunately, treatment options for these disorders are severely lacking. Challenges in designing therapeutics for use during pregnancy involve selectively delivering payloads to the placenta while protecting the fetus from potential toxic side effects. Nanomedicine holds great promise in overcoming these barriers; the versatile and modular nature of nanocarriers, including prolonged circulation times, intracellular delivery, and organ-specific targeting, can control how therapeutics interact with the placenta. In this review, nanomedicine strategies are discussed to treat and diagnose placental disorders with an emphasis on understanding the unique pathophysiology behind each of these diseases. Finally, prior study of the pathophysiologic mechanisms underlying these placental disorders has revealed novel disease targets. These targets are highlighted here to motivate the rational design of precision nanocarriers to improve therapeutic options for placental disorders.
Collapse
Affiliation(s)
- Hannah C. Geisler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Hannah C. Safford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19014, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| |
Collapse
|
7
|
Cheng G, Qu R, Song G, Li X, Zhang F, Wang J, Li W, Wen J, Xiao Y, Wei Y, Zhang W, Yi X, Li S, Ding J, Zhang Y. Association of ambient temperature and diurnal temperature range with the outcome of in vitro fertilization in women from Hubei, China: A retrospective cohort study. ENVIRONMENTAL RESEARCH 2024; 263:120072. [PMID: 39341537 DOI: 10.1016/j.envres.2024.120072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/08/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
OBJECTIVES To examine the relationship between ambient temperature and DTR and pregnancy outcomes in vitro fertilization/intracytoplasmic monosperm injection and embryo transfer (IVF/ICSI-ET) women. METHODS The study included 5264 women who were treated with IVF/ICSI-ET at two centers in Hubei province from 2017 to 2022. The daily mean, daily maximum, and daily minimum temperatures at the subjects' home addresses were extracted, and DTR values were calculated based on latter two. The associations between ambient temperature and DTR with clinical pregnancy and live birth rates were assessed using multivariate logistic regression models adjusted for covariates. Subgroup analyses were also conducted to explore potential modifiers. RESULT High/low temperatures as well as a larger DTR had a significant effect on pregnancy outcomes in fresh cycles, but not in FET cycles. Specifically, hot weather exposure to high temperatures was associated with reduced clinical pregnancy rates: Period 4 (embryo transfer to serum HCG testing) (aOR = 0.873, 95%CI: 0.763-0.999). Ambient temperature in cold weather was positively associated with live birth rate: Period 2 (Gn initiation to oocyte retrieval) (aOR = 1.082, 95% CI: 1.01-1.170), Period 3 (oocyte retrieval to embryo transfer) (aOR = 1.111, 95% CI: 1.019-1.212), Period 4 (aOR = 1.134, 95% CI: 1.028-1.252), and Period 7 (85 days prior to oocyte retrieval to serum hCG testing) (aOR = 1.105, 95% CI: 1.007-1.212). For DTR, exposure to larger DTR (Q3) at Period 2, Period 3, and Period 6 (Gn initiation to embryo transfer) reduces clinical pregnancy and live birth rates compared with Q1. Subgroup analyses revealed susceptibility profiles across age groups and residential address populations in different sensitivity windows. CONCLUSION Our study shows that exposure to hot and cold weather and higher DTR reduces clinical pregnancy rates and live birth rates in women undergoing fresh embryo transfer, but has no significant effect on FET cycles.
Collapse
Affiliation(s)
- Guan Cheng
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Rui Qu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Ge Song
- Hubei Key Laboratory of Quantitative Remote Sensing of Land and Atmosphere, School of Remote Sensing and Information Engineering, Wuhan University, Hubei, 430000, China
| | - Xing Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Feng Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Jingxuan Wang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Wenzhu Li
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Jiahui Wen
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Yanfei Xiao
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Yiqiu Wei
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Weiqian Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Xiaofang Yi
- Department of Reproductive Medicine, Renhe Hospital, Three Gorges University, Yichang, 443000, China
| | - Siwei Li
- Hubei Key Laboratory of Quantitative Remote Sensing of Land and Atmosphere, School of Remote Sensing and Information Engineering, Wuhan University, Hubei, 430000, China; Hubei Luojia Laboratory, Wuhan University, Hubei, 430000, China; State Key Laboratory of Information Engineering in Surveying, Mapping and Remote Sensing, Wuhan University, Hubei, 430000, China
| | - Jinli Ding
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Yan Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
Collapse
|
8
|
Nagashima M, Takeda Y, Saitoh S, Sabrina S, Araki A, Nagase S, Asao H. A loss of tuning of both pro-coagulant and inflammatory responses in monocytes in patients with preeclampsia. J Reprod Immunol 2024; 166:104334. [PMID: 39332076 DOI: 10.1016/j.jri.2024.104334] [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: 06/14/2024] [Revised: 08/30/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
An imbalance between pro- and anti-angiogenesis is one of the leading causes of preeclampsia (PE). Monocytes, known as regulators of angiogenesis during immune responses, cooperate with platelets, but the specifics of these responses during pregnancy remain unclear. In this study, we investigated the relationship between pro-coagulant responses on monocytes [platelet activation marker CD61 as a monocyte-platelet aggregate (MPA), tissue factor (CD142), etc.], inflammatory responses [soluble CD40 ligand (sCD40L), soluble suppression of tumorigenesis-2 (sST2), etc.], and the balance of angiogenesis [soluble Fms-related receptor tyrosine kinase 1/placental growth factor (sFlt-1/PLGF) ratio]. In PE, markers of pro-coagulant and inflammatory responses were higher than those in normal pregnancy (NP). Interestingly, in NP, these markers harmonized with the sFlt-1/PLGF ratio, but not in PE. Furthermore, ex vivo examinations showed that upregulation of CD142 induced by additional platelet activation with adenosine diphosphate was diminished in PE. Conversely, low-dose aspirin, which is used as a preventive treatment for PE, could inhibit the increase of CD61 and sST2 under inflammatory stimuli and platelet activation in NP but not in PE. These results indicate that monocytes in PE upregulate basal activity and lose responsiveness to stimulation. The elevation of pro-coagulant and inflammatory responses may be mitigated by prophylaxis with low-dose aspirin. Therefore, the findings suggesting a loss of tuning of both pro-coagulant and inflammatory responses on monocytes help in understanding the pathology of PE. The harmonization between pro-coagulant responses, inflammatory responses, and angiogenesis may serve as useful indicators for the prediction and preventive treatment of PE.
Collapse
Affiliation(s)
- Mikako Nagashima
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan; Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Yuji Takeda
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan.
| | - Shinichi Saitoh
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Saima Sabrina
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Akemi Araki
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Satoru Nagase
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Hironobu Asao
- Department of Immunology, Yamagata University Faculty of Medicine, Yamagata, Japan
| |
Collapse
|
9
|
Ovadia M, Gluska H, Cohen G, Schreiber H, Biron-Shental T, Kovo M, Shechter-Maor G. Does decreased fetal growth estimation in the appropriate for gestational age range affect delivery outcomes? Arch Gynecol Obstet 2024; 310:1461-1465. [PMID: 38411630 DOI: 10.1007/s00404-024-07432-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/12/2024] [Indexed: 02/28/2024]
Abstract
PURPOSE To study the effect of decreased estimated fetal weight (EFW) percentiles in appropriate for gestational age fetuses. METHODS This retrospective cohort study included women who had second and third trimester ultrasound examinations. Delivery and neonatal outcomes of pregnancies with decreased EFW of ≥ 30 percentiles in EFW between ultrasound examinations (decreased growth group) and those without such a decrease (control group) were compared. Deliveries with EFW or birthweight below the 10th percentile were excluded. RESULTS Among 1610 deliveries, 57 were in the decreased growth group and 1553 in the control group. Maternal characteristics did not differ between the groups except for higher rate of nulliparity in the decreased growth group. We found similar rates of Category II/III monitoring, cesarean deliveries due to non-reassuring fetal heart rate and adverse neonatal outcomes. Neonatal birthweight was lower in the decreased growth group as compared to controls. CONCLUSIONS This study did not find association between the group of appropriate for gestational age fetuses with decreased growth, with adverse outcomes.
Collapse
Affiliation(s)
- Michal Ovadia
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Affiliated with School of Medicine, Tel Aviv University, 59 Tchernichovsky St, 44281, Kfar Saba, Israel
| | - Hadar Gluska
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Affiliated with School of Medicine, Tel Aviv University, 59 Tchernichovsky St, 44281, Kfar Saba, Israel
| | - Gal Cohen
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Affiliated with School of Medicine, Tel Aviv University, 59 Tchernichovsky St, 44281, Kfar Saba, Israel
| | - Hanoch Schreiber
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Affiliated with School of Medicine, Tel Aviv University, 59 Tchernichovsky St, 44281, Kfar Saba, Israel
| | - Tal Biron-Shental
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Affiliated with School of Medicine, Tel Aviv University, 59 Tchernichovsky St, 44281, Kfar Saba, Israel
| | - Michal Kovo
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Affiliated with School of Medicine, Tel Aviv University, 59 Tchernichovsky St, 44281, Kfar Saba, Israel
| | - Gil Shechter-Maor
- Department of Obstetrics and Gynecology, Meir Medical Center, Kfar Saba, Affiliated with School of Medicine, Tel Aviv University, 59 Tchernichovsky St, 44281, Kfar Saba, Israel.
| |
Collapse
|
10
|
Turan OM, Liang Y, Kelley B, Turan S, Pepe GJ, Albrecht ED. B-flow/spatiotemporal image correlation M-mode ultrasound provides novel method to quantify spiral artery remodeling during normal human pregnancy. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 64:322-329. [PMID: 38477161 PMCID: PMC11371540 DOI: 10.1002/uog.27636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
OBJECTIVES During human pregnancy, placental extravillous trophoblasts replace vascular smooth muscle and elastic tissue within the walls of the uterine spiral arteries, thereby remodeling them into distensible low-resistance vessels to promote placental perfusion. The present study determined whether B-flow/spatiotemporal image correlation (STIC) M-mode ultrasonography provides an in-vivo imaging method able to digitally quantify spiral artery luminal distensibility as a physiological index of spiral artery remodeling during the advancing stages of normal human pregnancy. METHODS A prospective, longitudinal, observational study was conducted to quantify spiral artery distensibility (i.e. vessel luminal diameter at systole minus diameter at diastole) by B-flow/STIC M-mode ultrasonography during the first, second and third trimesters in 290 women exhibiting a normal pregnancy. Maternal serum levels of placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1), growth factors that modulate important events in spiral artery remodeling, were quantified in a subset of the women in the first, second and third trimesters of pregnancy. RESULTS Median (interquartile range (IQR)) spiral artery distensibility increased progressively between the first (0.17 (0.14-0.21) cm), second (0.23 (0.18-0.28) cm) and third (0.26 (0.21-0.35) cm) trimesters of pregnancy (P < 0.0001 for all). Median (IQR) spiral artery volume flow increased progressively between the first (2.49 (1.38-4.99) mL/cardiac cycle), second (3.86 (2.06-6.91) mL/cardiac cycle) and third (7.79 (3.83-14.98) mL/cardiac cycle) trimesters (P < 0.001 for all). In accordance with the elevation in spiral artery distensibility, the median (IQR) ratio of serum PlGF/sFlt-1 × 103 levels increased between the first (7.2 (4.5-10.0)), second (22.7 (18.6-42.2)) and third (56.2 (41.9-92.5)) trimesters (P < 0.001 for all). CONCLUSIONS The present study shows that B-flow/STIC M-mode ultrasonography provides an in-vivo imaging technology to quantify digitally the structural and physiological expansion of the walls of the spiral arteries during the cardiac cycle as a consequence of their transformation into compliant vessels during advancing stages of normal human pregnancy. © 2024 International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- O M Turan
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Y Liang
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - B Kelley
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - S Turan
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - G J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - E D Albrecht
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Scher MS. Neonatal Encephalopathy is a Complex Phenotype Representing Reproductive and Pregnancy Exposome Effects on the Maternal-Placental-Fetal Triad. Clin Perinatol 2024; 51:535-550. [PMID: 39095094 DOI: 10.1016/j.clp.2024.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Reproductive, pregnancy, and placental exposomes influence the fetal neural exposome through toxic stressor interplay, impairing the maternal-placental-fetal (MPF) triad. Neonatal encephalopathy represents different clinical presentations based on complex time-dependent etiopathogenetic mechanisms including hypoxia-ischemia that challenge diagnosis and prognosis. Reproductive, pregnancy, and placental exposomes impair the fetal neural exposome through toxic stressor interplay within the MPF triad. Long intervals often separate disease onset from phenotype. Interdisciplinary fetal-neonatal neurology training, practice, and research closes this knowledge gap. Maintaining reproductive health preserves MPF triad health with life-course benefits.
Collapse
Affiliation(s)
- Mark S Scher
- Division of Pediatric Neurology, Department of Pediatrics, Fetal/Neonatal Neurology Program, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital/ MacDonald Hospital for Women, University Hospitals Cleveland Medical Center, 22315 Canterbury Lane, Shaker Heights, OH 44122, USA.
| |
Collapse
|
12
|
Roberts D, Aisagbonhi O, Parast MM. Incorporating placental pathology into clinical care and research. Trends Mol Med 2024:S1471-4914(24)00216-8. [PMID: 39299838 DOI: 10.1016/j.molmed.2024.08.002] [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: 06/19/2024] [Revised: 08/06/2024] [Accepted: 08/06/2024] [Indexed: 09/22/2024]
Abstract
Despite recent standardization of placental evaluation and establishment of criteria for diagnosis of major patterns of placental injury, placental pathological examination remains undervalued and under-utilized. The placenta can harbor a significant amount of information relevant to both the pregnant person and offspring. Placental pathology can also provide a significant context for pathophysiological study of adverse pregnancy outcomes, helping to optimally subcategorize the 'great obstetric syndromes' of pre-eclampsia (PE), spontaneous preterm birth (sPTB), and fetal growth restriction (FGR), and to identify causes of stillbirth. We hereby propose that placental evaluation should be incorporated into routine delivery of obstetric and neonatal care, and further suggest that its integration into clinical, translational, and basic research could significantly advance our understanding of pregnancy complications and adverse neonatal outcomes.
Collapse
Affiliation(s)
- Drucilla Roberts
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Omonigho Aisagbonhi
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Mana M Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA.
| |
Collapse
|
13
|
Doria-Borrell P, Pérez-García V. Understanding the intersection between placental development and cancer: Lessons from the tumor suppressor BAP1. Commun Biol 2024; 7:1053. [PMID: 39191942 DOI: 10.1038/s42003-024-06689-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
The placenta, a pivotal organ in mammalian reproduction, allows nutrient exchange and hormonal signaling between the mother and the developing fetus. Understanding its molecular intricacies is essential for deciphering normal embryonic development and pathological conditions such as tumorigenesis. Here, we explore the multifaceted role of the tumor suppressor BRCA1-associated protein 1 (BAP1) in cancer and placentation. Initially recognized for its tumor-suppressive properties, BAP1 has emerged as a key regulator at the intersection of tumorigenesis and placental development. BAP1 influences crucial cellular processes such as cell death, proliferation, metabolism, and response to hypoxic conditions. By integrating insights from tumor and developmental biology, we illuminate the complex molecular pathways orchestrated by BAP1. This perspective highlights BAP1's significant impact on both cancer and placental development, and suggests novel therapeutic strategies that could improve outcomes for pregnancy disorders and cancer.
Collapse
Affiliation(s)
| | - Vicente Pérez-García
- Centro de Investigación Príncipe Felipe, Valencia, Spain.
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain.
| |
Collapse
|
14
|
Shukla V, Moreno-Irusta A, Varberg KM, Kuna M, Iqbal K, Galligos AM, Aplin JD, Choudhury RH, Okae H, Arima T, Soares MJ. NOTUM-MEDIATED WNT SILENCING DRIVES EXTRAVILLOUS TROPHOBLAST CELL LINEAGE DEVELOPMENT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.13.579974. [PMID: 38405745 PMCID: PMC10888853 DOI: 10.1101/2024.02.13.579974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Trophoblast stem (TS) cells have the unique capacity to differentiate into specialized cell types, including extravillous trophoblast (EVT) cells. EVT cells invade into and transform the uterus where they act to remodel the vasculature facilitating the redirection of maternal nutrients to the developing fetus. Disruptions in EVT cell development and function are at the core of pregnancy-related disease. WNT-activated signal transduction is a conserved regulator of morphogenesis of many organ systems, including the placenta. In human TS cells, activation of canonical WNT signaling is critical for maintenance of the TS cell stem state and its downregulation accompanies EVT cell differentiation. We show that aberrant WNT signaling undermines EVT cell differentiation. Notum, palmitoleoyl-protein carboxylesterase (NOTUM), a negative regulator of canonical WNT signaling, was prominently expressed in first trimester EVT cells developing in situ and upregulated in EVT cells derived from human TS cells. Furthermore, NOTUM was required for optimal human TS cell differentiation to EVT cells. Activation of NOTUM in EVT cells is driven, at least in part, by endothelial PAS domain 1 (also called hypoxia-inducible factor 2 alpha). Collectively, our findings indicate that canonical WNT signaling is essential for maintenance of human trophoblast cell stemness and regulation of human TS cell differentiation. Downregulation of canonical WNT signaling via the actions of NOTUM is required for optimal EVT cell differentiation.
Collapse
Affiliation(s)
- Vinay Shukla
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Ayelen Moreno-Irusta
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Kaela M. Varberg
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Marija Kuna
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Khursheed Iqbal
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - Anna M. Galligos
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
| | - John D. Aplin
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, The University of Manchester, Manchester M13 9WL, United Kingdom
- Manchester Academic Health Sciences Centre, St Mary’s Hospital, University of Manchester, Manchester M13 9WL, United Kingdom
| | - Ruhul H. Choudhury
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, The University of Manchester, Manchester M13 9WL, United Kingdom
- Manchester Academic Health Sciences Centre, St Mary’s Hospital, University of Manchester, Manchester M13 9WL, United Kingdom
| | - Hiroaki Okae
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811 Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Michael J. Soares
- Institute for Reproductive and Developmental Sciences, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS
- Center for Perinatal Research, Children’s Research Institute, Children’s Mercy, Kansas City, MO
- Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
15
|
Xia S, Ye Y, Liu J, Qiu H, Lin M, He Z, Huang L, Wang M, Luo Y. The Role of MALAT1 in Regulating the Proangiogenic Functions, Invasion, and Migration of Trophoblasts in Selective Fetal Growth Restriction. Biomolecules 2024; 14:988. [PMID: 39199376 PMCID: PMC11352967 DOI: 10.3390/biom14080988] [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: 07/02/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
Epigenetic regulation is an important entry point to study the pathogenesis of selective fetal growth restriction (sFGR), and an understanding of the role of long noncoding RNAs (lncRNAs) in sFGR is lacking. Our study aimed to investigate the potential role of a lncRNA, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), in sFGR using molecular biology experiments and gain- or loss-of-function assays. We found that the levels of MALAT1, ERRγ, and HSD17B1 were downregulated and that of miR-424 was upregulated in the placental shares of the smaller twins. Moreover, angiogenesis was impaired in the placental share of the smaller fetus and MALAT1 could regulate the paracrine effects of trophoblasts on endothelium angiogenesis and proliferation by regulating miR-424. In trophoblasts, MALAT1 could competitively bind to miR-424 to regulate the expression of ERRγ and HSD17B1, thus regulating trophoblast invasion and migration. MALAT1 overexpression could decrease apoptosis and promote proliferation, alleviating cell damage induced by hypoxia. Taken together, the downregulation of MALAT1 can reduce the expression of ERRγ and HSD17B1 by competitively binding to miR-424, impairing the proangiogenic effect of trophoblasts, trophoblast invasion and migration, and the ability of trophoblasts to compensate for hypoxia, which may be involved in the pathogenesis of sFGR through various aspects.
Collapse
Affiliation(s)
- Shuting Xia
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Yingnan Ye
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Jialiu Liu
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Hanfei Qiu
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Minhuan Lin
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Zhiming He
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Linhuan Huang
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Malie Wang
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| | - Yanmin Luo
- Department of Obstetrics & Gynecology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; (S.X.); (Y.Y.); (J.L.); (H.Q.); (M.L.); (Z.H.); (L.H.); (M.W.)
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou 510080, China
| |
Collapse
|
16
|
Shen Y, Lian Y, Xiao L, Miu Y, Niu J, Cui Q. GPR124 promotes trophoblast proliferation, migration, and invasion and inhibits trophoblast cell apoptosis and inflammation via JNK and P38 MAPK pathways. J Cell Physiol 2024; 239:e31298. [PMID: 38764331 DOI: 10.1002/jcp.31298] [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: 01/02/2024] [Revised: 04/04/2024] [Accepted: 05/03/2024] [Indexed: 05/21/2024]
Abstract
Early-onset preeclampsia, which occurrs before 34 weeks of gestation, is the most dangerous classification of preeclampsia, which is a pregnancy-specific disease that causes 1% of maternal deaths. G protein-coupled receptor 124 (GPR124) is significantly expressed at various stages of the human reproductive process, particularly during embryogenesis and angiogenesis. Our prior investigation demonstrated a notable decrease in GPR124 expression in the placentas of patients with early-onset preeclampsia compared to that in normal pregnancy placentas. However, there is a lack of extensive investigation into the molecular processes that contribute to the role of GPR124 in placenta development. This study aimed to examine the mechanisms by which GPR124 affects the occurrence of early-onset preeclampsia and its function in trophoblast. Proliferative, invasive, migratory, apoptotic, and inflammatory processes were identified in GPR124 knockdown, GPR124 overexpression, and normal HTR8/SVneo cells. The mechanism of GPR124-mediated cell function in GPR124 knockdown HTR8/SVneo cells was examined using inhibitors of the JNK or P38 MAPK pathway. Downregulation of GPR124 was found to significantly inhibit proliferation, invasion and migration, and promote apoptosis of HTR8/SVneo cells when compared to the control and GPR124 overexpression groups. This observation is consistent with the pathological characteristics of preeclampsia. In addition, GPR124 overexpression inhibits the secretion of pro-inflammatory cytokines interleukin (IL)-8 and interferon-γ (IFN-γ) while enhancing the secretion of the anti-inflammatory cytokine interleukin (IL)-4. Furthermore, GPR124 suppresses the activation of P-JNK and P-P38 within the JNK/P38 MAPK pathway. The invasion, apoptosis, and inflammation mediated by GPR124 were partially restored by suppressing the JNK and P38 MAPK pathways in HTR8/SVneo cells. GPR124 plays a crucial role in regulating trophoblast proliferation, invasion, migration, apoptosis, and inflammation via the JNK and P38 MAPK pathways. Furthermore, the effect of GPR124 on trophoblast suggests its involvement in the pathogenesis of early-onset preeclampsia.
Collapse
Affiliation(s)
- Yan Shen
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Cheeloo College of Medicine, Shandong University, shenzhen, Guangdong, China
- Department of Obstetrics, Shandong Provincial Maternal and Child Health Care Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, China
| | - Yan Lian
- Department of Obstetrics, Shandong Provincial Maternal and Child Health Care Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, China
| | - Li Xiao
- Department of Obstetrics, Shandong Provincial Maternal and Child Health Care Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, China
| | - Yaya Miu
- Department of Obstetrics, Shandong Provincial Maternal and Child Health Care Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, China
| | - Jianmin Niu
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Cheeloo College of Medicine, Shandong University, shenzhen, Guangdong, China
- Department of Obstetrics, The Eight Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Qingyu Cui
- The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
17
|
Parker J, Hofstee P, Brennecke S. Prevention of Pregnancy Complications Using a Multimodal Lifestyle, Screening, and Medical Model. J Clin Med 2024; 13:4344. [PMID: 39124610 PMCID: PMC11313446 DOI: 10.3390/jcm13154344] [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: 06/19/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Prevention of pregnancy complications related to the "great obstetrical syndromes" (preeclampsia, fetal growth restriction, spontaneous preterm labor, and stillbirth) is a global research and clinical management priority. These syndromes share many common pathophysiological mechanisms that may contribute to altered placental development and function. The resulting adverse pregnancy outcomes are associated with increased maternal and perinatal morbidity and mortality and increased post-partum risk of cardiometabolic disease. Maternal nutritional and environmental factors are known to play a significant role in altering bidirectional communication between fetal-derived trophoblast cells and maternal decidual cells and contribute to abnormal placentation. As a result, lifestyle-based interventions have increasingly been recommended before, during, and after pregnancy, in order to reduce maternal and perinatal morbidity and mortality and decrease long-term risk. Antenatal screening strategies have been developed following extensive studies in diverse populations. Multivariate preeclampsia screening using a combination of maternal, biophysical, and serum biochemical markers is recommended at 11-14 weeks' gestation and can be performed at the same time as the first-trimester ultrasound and blood tests. Women identified as high-risk can be offered prophylactic low dose aspirin and monitored with angiogenic factor assessment from 22 weeks' gestation, in combination with clinical assessment, serum biochemistry, and ultrasound. Lifestyle factors can be reassessed during counseling related to antenatal screening interventions. The integration of lifestyle interventions, pregnancy screening, and medical management represents a conceptual advance in pregnancy care that has the potential to significantly reduce pregnancy complications and associated later life cardiometabolic adverse outcomes.
Collapse
Affiliation(s)
- Jim Parker
- School of Medicine, University of Wollongong, Wollongong 2522, Australia;
| | - Pierre Hofstee
- School of Medicine, University of Wollongong, Wollongong 2522, Australia;
- Tweed Hospital, Northern New South Wales Local Health District, Tweed Heads 2485, Australia
| | - Shaun Brennecke
- Department of Maternal-Fetal Medicine, Pregnancy Research Centre, The Royal Women’s Hospital, Melbourne 3052, Australia;
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne 3052, Australia
| |
Collapse
|
18
|
Scher MS. Interdisciplinary fetal-neonatal neurology training improves brain health across the lifespan. Front Neurol 2024; 15:1411987. [PMID: 39026582 PMCID: PMC11254674 DOI: 10.3389/fneur.2024.1411987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
Integrated fetal, neonatal, and pediatric training constitute an interdisciplinary fetal-neonatal neurology (FNN) program. A dynamic neural exposome concept strengthens curriculum content. Trainees participate in mentoring committee selection for guidance during a proposed two-year program. Prenatal to postnatal clinical learning re-enforces early toxic stressor interplay that influences gene-environment interactions. Maternal-placental-fetal triad, neonatal, or childhood diseases require diagnostic and therapeutic decisions during the first 1,000 days when 80 % of neural connections contribute to life-course phenotypic expression. Pediatric follow-up through 3 years adjusts to gestational ages of preterm survivors. Cumulative reproductive, pregnancy, pediatric and adult exposome effects require educational experiences that emphasize a principle-to-practice approach to a brain capital strategy across the lifespan. More rigorous training during fetal, neonatal, and pediatric rotations will be offered to full time trainees. Adult neurology residents, medical students, and trainees from diverse disciplines will learn essential topics during time-limited rotations. Curriculum content will require periodic re-assessments using educational science standards that maintain competence while promoting creative and collaborative problem-solving. Continued career-long learning by FNN graduates will strengthen shared healthcare decisions by all stakeholders. Recognition of adaptive or maladaptive neuroplasticity mechanisms requires analytic skills that identify phenotypes associated with disease pathways. Developmental origins and life-course concepts emphasize brain health across the developmental-aging continuum, applicable to interdisciplinary research collaborations. Social determinants of health recognize diversity, equity, and inclusion priorities with each neurological intervention, particularly for those challenged with disparities. Diagnostic and therapeutic strategies must address resource challenges particularly throughout the Global South to effectively lower the worldwide burden of neurologic disease. Sustainable development goals proposed by the World Health Organization offer universally applicable guidelines in response to ongoing global and regional polycrises. Gender, race, ethnicity, and socio-economic equality promote effective preventive, rescue and reparative neuroprotective interventions. Global synergistic efforts can be enhanced by establishing leadership within academic teaching hubs in FNN training to assist with structure and guidance for smaller healthcare facilities in each community that will improve practice, education and research objectives. Reduced mortality with an improved quality of life must prioritize maternal-pediatric health and well-being to sustain brain health across each lifespan with transgenerational benefits.
Collapse
Affiliation(s)
- Mark S. Scher
- Department of Pediatrics and Neurology, Division of Pediatric Neurology, Fetal/Neonatal Neurology Program, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
19
|
Bhardwaj B, Singh S, Kumar S, Nandy S, Kumari A, Khardenavis N. Comparative study of difference in anatomy & histopathology of placenta & umbilical cord in natural pregnancy vs. IVF pregnancy & it's impact on fetal & maternal outcome. Eur J Obstet Gynecol Reprod Biol 2024; 298:204-207. [PMID: 38796875 DOI: 10.1016/j.ejogrb.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
AIM AND OBJECTIVES Comparison of naturally conceived pregnancy with IVFET pregnancy for feto-maternal outcome and morphology and histopathology of placenta & umbilical cord. METHODS 100 pregnant women were divided into 2 subsets of spontaneous pregnancy group (n = 50) and the IVFET pregnancy group (n = 50).The two groups were compared for Maternal age, parity, maternal weight gain, prepregnancy maternal BMI, gestational age, birth weight of baby, placental weight, placenta and umbilical cord cross sections, insertion site of the umbilical cord, and length of the umbilical cord. INCLUSION CRITERIA Patients registered at ANC OPD/ART centre of our institute and subsequently reporting to maternity ward/ labor room for delivery at our centre. EXCLUSION CRITERIA The pregnancies conceived after ART outside our institute, multifetal pregnancies. Study duration: 01 year Results: Our study revealed that spontaneous pregnancy group had less antenatal co-morbidities with more number of term vaginal deliveries and less intrapartum and neonatal complications compared to IVFET pregnancy women (p < 0.05). CONCLUSIONS Assisted reproductive technologies have an impact on placental growth and function in pregnancy. The occurrence of placental abnormalities were the most significant and pertinent finding in the IVF-ET placentas. On histopathological examination maternal vascular malperfusion and concomitant anomalies of the umbilical cord were most noticeable findings.
Collapse
|
20
|
Geisler HC, Ghalsasi AA, Safford HC, Swingle KL, Thatte AS, Mukalel AJ, Gong N, Hamilton AG, Han EL, Nachod BE, Padilla MS, Mitchell MJ. EGFR-targeted ionizable lipid nanoparticles enhance in vivo mRNA delivery to the placenta. J Control Release 2024; 371:455-469. [PMID: 38789090 PMCID: PMC11259947 DOI: 10.1016/j.jconrel.2024.05.036] [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: 11/30/2023] [Revised: 05/15/2024] [Accepted: 05/19/2024] [Indexed: 05/26/2024]
Abstract
The full potential of ionizable lipid nanoparticles (LNPs) as an in vivo nucleic acid delivery platform has not yet been realized given that LNPs primarily accumulate in the liver following systemic administration, limiting their success to liver-centric conditions. The engineering of LNPs with antibody targeting moieties can enable extrahepatic tropism by facilitating site-specific LNP tethering and driving preferential LNP uptake into receptor-expressing cell types via receptor-mediated endocytosis. Obstetric conditions stemming from placental dysfunction, such as preeclampsia, are characterized by overexpression of cellular receptors, including the epidermal growth factor receptor (EGFR), making targeted LNP platforms an exciting potential treatment strategy for placental dysfunction during pregnancy. Herein, an EGFR antibody-conjugated LNP (aEGFR-LNP) platform was developed by engineering LNPs with increasing densities of antibody functionalization. aEGFR-LNPs were screened in vitro in immortalized placental trophoblasts and in vivo in non-pregnant and pregnant mice and compared to non-targeted formulations for extrahepatic, antibody-targeted mRNA LNP delivery to the placenta. Our top performing LNP with an intermediate density of antibody functionalization (1:5 aEGFR-LNP) mediated a ∼twofold increase in mRNA delivery in murine placentas and a ∼twofold increase in LNP uptake in EGFR-expressing trophoblasts compared to non-targeted counterparts. These results demonstrate the potential of antibody-conjugated LNPs for achieving extrahepatic tropism, and the ability of aEGFR-LNPs in promoting mRNA delivery to EGFR-expressing cell types in the placenta.
Collapse
Affiliation(s)
- Hannah C Geisler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Aditi A Ghalsasi
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Hannah C Safford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Ajay S Thatte
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Alvin J Mukalel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Alex G Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Emily L Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Benjamin E Nachod
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Marshall S Padilla
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for RNA Innovation, Perelman School of Medicine, Philadelphia, PA, United States; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
21
|
Zhi Y, Zhang P, Luo Y, Sun Y, Li J, Zhang M, Li Y. CXC chemokine receptor type 5 may induce trophoblast dysfunction and participate in the processes of unexplained missed abortion, wherein p-ERK and interleukin-6 may be involved. Heliyon 2024; 10:e31465. [PMID: 38882363 PMCID: PMC11176800 DOI: 10.1016/j.heliyon.2024.e31465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 06/18/2024] Open
Abstract
Chemokines regulate the trophoblast dysfunction involved in the occurrence and development of pathological pregnancy, including missed abortions. In particular, CXC chemokine receptor type 5 mediates cell proliferation, migration, and inflammation; nonetheless, its role in missed abortions remains unclear. This study aimed to examine the expression of CXC chemokine receptor type 5 in missed abortions and to investigate the effects of CXC chemokine receptor type 5 on the biological behaviour of trophoblasts, as well as the underlying mechanisms. Our results indicated that CXC chemokine receptor type 5 was upregulated in the villi of women who experienced unexplained missed abortions, as compared with those who had normal pregnancies. CXC chemokine receptor type 5 inhibited the proliferation and migration of human first-trimester trophoblast/simian virus cells but promoted cell apoptosis. With respect to its mechanisms, CXC chemokine receptor type 5 activated the extracellular signal-regulated protein kinase 1/2 signalling pathway and upregulated the secretion of interleukin-6; however, it had no effect on the secretion of tumour necrosis factor-α. In conclusion, our findings suggest that CXC chemokine receptor type 5 induces trophoblast dysfunction and participates in the processes of unexplained missed abortions, wherein p-ERK and interleukin-6 may be involved.
Collapse
Affiliation(s)
- Yanan Zhi
- Department of Reproductive and Genetics, Hebei General Hospital, Shijiazhuang, Hebei, PR China
- Second Ward of Gynecology, Dingzhou People's Hospital, Baoding, Hebei, PR China
| | - Pingping Zhang
- Department of Reproductive and Genetics, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| | - Yan Luo
- Department of Reproductive and Genetics, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| | - Yanmei Sun
- Department of Reproductive and Genetics, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| | - Juan Li
- Department of Reproductive and Genetics, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| | - Mingming Zhang
- Hebei Key Laboratory of Metabolic Diseases, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| | - Yali Li
- Department of Reproductive and Genetics, Hebei General Hospital, Shijiazhuang, Hebei, PR China
| |
Collapse
|
22
|
Krog MC, Flachs EM, Kolte AM, de Jager W, Meyaard L, Christiansen OB, Steffensen R, Vomstein K, Garred P, Nielsen HS. Angiogenic factors and the lectin pathway of complement in women with secondary recurrent pregnancy loss. J Reprod Immunol 2024; 163:104221. [PMID: 38447288 DOI: 10.1016/j.jri.2024.104221] [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: 07/06/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
The poor remodeling of placental spiral arteries seen in preeclampsia is also discussed to contribute to recurrent pregnancy loss (RPL) preceded by abnormal angiogenesis and excessive complement activation. Low levels of Mannose-binding-lectin (MBL), a pattern recognition molecule (PRM) of the lectin pathway, have been found in women with RPL. We propose that pregnancy loss is connected to defective angiogenesis with reperfusion damage in the placenta and decreased levels of PRM in the lectin pathway in women with RPL. In this cohort study, we investigate the angiogenic factors and the lectin complement pathway in early pregnancy and their time-dependent relationship with pregnancy outcomes in 76 women with secondary RPL (sRPL) who have at least four prior pregnancy losses and a live birth. We evaluated levels of Angiopoietin-1 (Ang-1), Angiopoietin-2 (Ang-2), Vascular Endothelial Growth Factor (VEGF), soluble fms-like tyrosine kinase-1 (sFlt-1), and the PRMs, MBL, ficolin-1, -2, -3 and an additional soluble PRM, Pentraxin-3, during the 5th, 6th, and 7th gestational weeks. Our results showed that, compared to live births, pregnancies that ended in loss were associated with elevated VEGF levels and decreased levels of the Ang-2/Ang-1 ratio. Also, increasing levels of ficolin-2 were significantly associated with pregnancy loss, with MBL showing no association. Our research suggests that women with sRPL may have inadequate placentation with impaired angiogenesis in pregnancies ending in a loss.
Collapse
Affiliation(s)
- M C Krog
- The Recurrent Pregnancy Loss Unit, the Capital Region, Copenhagen University Hospitals, Rigshospitalet and Hvidovre Hospital, Kettegård Alle 30, Hvidovre 2650, Denmark; The Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, Copenhagen 2100, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Blegdamsvej 3B, Copenhagen 2200, Denmark.
| | - E M Flachs
- The Department of Occupational and Environmental Medicine, Copenhagen University Hospital, Bispebjerg Hospital, Bispebjerg Bakke 23F, Copenhagen 2400, Denmark
| | - A M Kolte
- The Recurrent Pregnancy Loss Unit, the Capital Region, Copenhagen University Hospitals, Rigshospitalet and Hvidovre Hospital, Kettegård Alle 30, Hvidovre 2650, Denmark
| | - W de Jager
- Multiplex Core Facility, Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands
| | - L Meyaard
- Multiplex Core Facility, Laboratory of Translational Immunology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht 3584 CX, the Netherlands
| | - O B Christiansen
- Centre for Recurrent Pregnancy Loss of Western Denmark, Department of Obstetrics and Gynecology, Aalborg University Hospital, Reberbansgade 15, Aalborg 9000, Denmark; Department of Clinical Medicine, Aalborg University, Søndre Skovvej 15, Aalborg 9000, Denmark
| | - R Steffensen
- Department of Clinical Immunology, Aalborg University Hospital, Urbansgade 32, Aalborg 9000, Denmark
| | - K Vomstein
- The Recurrent Pregnancy Loss Unit, the Capital Region, Copenhagen University Hospitals, Rigshospitalet and Hvidovre Hospital, Kettegård Alle 30, Hvidovre 2650, Denmark; Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre Hospital, Kettegård Alle 30, Hvidovre 2650, Denmark
| | - P Garred
- The Department of Clinical Immunology, Copenhagen University Hospital, Rigshospitalet, Blegdamsvej 9, Copenhagen 2100, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Blegdamsvej 3B, Copenhagen 2200, Denmark; The Laboratory of Molecular Medicine, Department of Clinical Immunology, Section 7631, Copenhagen University Hospital, Rigshospitalet, Ole Maaløesvej 26, Copenhagen 2200, Denmark
| | - H S Nielsen
- The Recurrent Pregnancy Loss Unit, the Capital Region, Copenhagen University Hospitals, Rigshospitalet and Hvidovre Hospital, Kettegård Alle 30, Hvidovre 2650, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Blegdamsvej 3B, Copenhagen 2200, Denmark; Department of Obstetrics and Gynecology, Copenhagen University Hospital, Hvidovre Hospital, Kettegård Alle 30, Hvidovre 2650, Denmark
| |
Collapse
|
23
|
Hong J, Crawford K, Cavanagh E, Clifton V, Kumar S. Prediction of preterm birth in women with fetal growth restriction - Is the weekly change in sFlt-1/PlGF ratio or PlGF levels useful? Acta Obstet Gynecol Scand 2024; 103:1112-1119. [PMID: 38483020 PMCID: PMC11103152 DOI: 10.1111/aogs.14831] [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: 11/30/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 05/21/2024]
Abstract
INTRODUCTION To assess the rate of change in soluble fms-like tyrosine kinase-1/placental growth factor (sFlt-1/PlGF) ratio and PlGF levels per week compared to a single sFlt-1/PlGF ratio or PlGF level to predict preterm birth for pregnancies complicated by fetal growth restriction. MATERIAL AND METHODS A prospective cohort study of pregnancies complicated by isolated fetal growth restriction. Maternal serum PlGF levels and the sFlt-1/PlGF ratio were measured at 4-weekly intervals from recruitment to delivery. We investigated the utility of PlGF levels, sFlt-1/PlGF ratio, change in PlGF levels per week or sFlt-1/PlGF ratio per week. Cox-proportional hazard models and Harrell's C concordance statistic were used to evaluate the effect of biomarkers on time to preterm birth. RESULTS The total study cohort was 158 pregnancies comprising 91 (57.6%) with fetal growth restriction and 67 (42.4%) with appropriate for gestational age controls. In the fetal growth restriction cohort, sFlt-1/PlGF ratio and PlGF levels significantly affected time to preterm birth (Harrell's C: 0.85-0.76). The rate of increase per week of the sFlt-1/PlGF ratio (hazard ratio [HR] 3.91, 95% confidence interval [CI]: 1.39-10.99, p = 0.01, Harrell's C: 0.74) was positively associated with preterm birth but change in PlGF levels per week was not (HR 0.65, 95% CI: 0.25-1.67, p = 0.37, Harrell's C: 0.68). CONCLUSIONS Both a high sFlt-1/PlGF ratio and low PlGF levels are predictive of preterm birth in women with fetal growth restriction. Although the rate of increase of the sFlt-1/PlGF ratio predicts preterm birth, it is not superior to either a single elevated sFlt-1/PlGF ratio or low PlGF level.
Collapse
Affiliation(s)
- Jesrine Hong
- Mater Research InstituteUniversity of QueenslandSouth BrisbaneQueenslandAustralia
- Faculty of MedicineThe University of QueenslandHerstonQueenslandAustralia
- Department of Obstetrics and Gynecology, Faculty of MedicineUniversiti MalayaKuala LumpurMalaysia
| | - Kylie Crawford
- Mater Research InstituteUniversity of QueenslandSouth BrisbaneQueenslandAustralia
- Faculty of MedicineThe University of QueenslandHerstonQueenslandAustralia
| | - Erika Cavanagh
- Mater Research InstituteUniversity of QueenslandSouth BrisbaneQueenslandAustralia
| | - Vicki Clifton
- Mater Research InstituteUniversity of QueenslandSouth BrisbaneQueenslandAustralia
| | - Sailesh Kumar
- Mater Research InstituteUniversity of QueenslandSouth BrisbaneQueenslandAustralia
- Faculty of MedicineThe University of QueenslandHerstonQueenslandAustralia
- NHMRC Centre for Research Excellence in Stillbirth, Mater Research InstituteUniversity of QueenslandBrisbaneQueenslandAustralia
| |
Collapse
|
24
|
Bacon SJ, Zhu Y, Ghosh P. Early spiral arteriole remodeling in the uterine-placental interface: A rat model. J Anat 2024; 244:1054-1066. [PMID: 38288680 PMCID: PMC11095304 DOI: 10.1111/joa.14019] [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: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 05/16/2024] Open
Abstract
The mammalian placenta's interface with the parent is a richly vascularized tissue whose development relies upon communication between many different cell types within the uterine microenvironment. The uterine blood vessels of the interface are reshaped during pregnancy into wide-bore, flaccid vessels that convey parental blood to the exchange region of the placenta. Invasive trophoblast as well as parental uterine macrophages and Natural Killer cells are involved in the stepwise remodeling of these vessels and their respective contributions to this crucial process are still being delineated. However, the earliest steps in arteriole remodeling are understudied as they are difficult to study in humans, and other species lack the deep trophoblast invasion that is so prominent a feature of placentation in humans. Here, we further characterize the rat, with deep hemochorial placentation akin to humans, as a model system in which to tease apart the earliest, relatively understudied events in spiral arteriole remodeling. We show that the rat uterine-placental interface increases in size and vascularity rapidly, before trophoblast invasion. The remodeling stages in the arterioles of the rat uterine-placental interface follow a sequence of anatomical changes similar to those in humans, and there are changes to the arterioles' muscular tunica media prior to the marked influx of immune cells. The rat is a tractable model in which to better understand the cell/cell interactions occurring in vivo in an intact tissue microenvironment over time.
Collapse
Affiliation(s)
- Sarah J Bacon
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Yuxi Zhu
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| | - Priyanjali Ghosh
- Department of Biological Sciences, Mount Holyoke College, South Hadley, Massachusetts, USA
| |
Collapse
|
25
|
Liu Q, Tan L, Yuan L, Chen X, Li F, He J, Gao R. Subacute exposure to DEHP leads to impaired decidual reaction and exacerbates the risk of early miscarriage in mice. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:331-341. [PMID: 38763769 PMCID: PMC11348695 DOI: 10.3724/zdxbyxb-2023-0583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/21/2024] [Indexed: 05/21/2024]
Abstract
OBJECTIVES To investigate the effect of subacute exposure of Di (2-ethylhexyl) phthalate (DEHP) on endometrial decidualization and early pregnancy miscarriage in mice. METHODS CD1 mice were orally administrated with 300 (low-dose group), 1000 (medium-dose group), or 3000 mg·kg-1·d-1 DEHP (1/10 LD50, high-dose group) for 28 days, respectively. An early natural pregnancy model and an artificially induced decidualization model were established. The uterine tissues were collected on D7 of natural pregnancy and D8 of artificially induced decidualization, respectively. The effects of a subacute exposure to DEHP on the decidualization of mice were detected by HE staining, Masson staining, TUNEL assay, and Western blotting. A model of spontaneous abortion was constructed in mice after subacute exposure to 300 mg·kg-1·d-1 DEHP, and the effect of impaired decidualization on pregnancy was investigated by observing the pregnancy outcome on the 10th day of gestation. RESULTS Compared with the control group, the conception rate was significantly decreased in the high-dose DEHP subacute exposure group (P<0.05). HE staining showed that, compared with the control group, the decidual stromal cells in the low- and medium-dose exposure groups were disorganized, the nuclei of the cells were irregular, the cytoplasmic staining was uneven, and the number of polymorphonuclear cells was significantly reduced. Masson staining showed that compared with the control group, the collagen fibers in the decidua region of the DEHP low-dose group and the medium-dose group were more distributed, more abundant and more disorderly. TUNEL assay showed increased apoptosis in the decidua area compared to the control group. Western blotting showed that the expression of BMP2, a marker molecule for endometrial decidualization, was significantly reduced (P<0.05 or P<0.01). The abortion rate and embryo resorption rate were increased, and the number of embryos, uterine wet weight, uterine area and placenta wet weight were decreased in DEHP low-dose group compared to the control group stimulated by mifepristone, an abortifacient drug (P<0.05 or P<0.01). CONCLUSIONS Subacute exposure to DEHP leads to impaired endometrial decidualization during early pregnancy and exacerbates the risk of adverse pregnancy outcomes in mice.
Collapse
Affiliation(s)
- Qiuju Liu
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, China.
| | - Liping Tan
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, China
| | - Liu Yuan
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, China
| | - Fangfang Li
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, China
| | - Junlin He
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, China
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health, Chongqing Medical University, Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
26
|
Ip PNP, Nguyen-Hoang L, Chaemsaithong P, Guo J, Wang X, Sahota DS, Chung JPW, Poon LCY. Ultrasonographic placental parameters at 11-13+6 weeks' gestation in the prediction of complications in pregnancy after assisted reproductive technology. Taiwan J Obstet Gynecol 2024; 63:341-349. [PMID: 38802197 DOI: 10.1016/j.tjog.2023.09.023] [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] [Accepted: 09/01/2023] [Indexed: 05/29/2024] Open
Abstract
OBJECTIVE To evaluate the performance of maternal factors, biophysical and biochemical markers at 11-13 + 6 weeks' gestation in the prediction of gestational diabetes mellitus with or without large for gestational age (GDM ± LGA) fetus and great obstetrical syndromes (GOS) among singleton pregnancy following in-vitro fertilisation (IVF)/embryo transfer (ET). MATERIALS AND METHODS A prospective cohort study was conducted between December 2017 and January 2020 including patients who underwent IVF/ET. Maternal mean arterial pressure (MAP), ultrasound markers including placental volume, vascularisation index (VI), flow index (FI) and vascularisation flow index (VFI), mean uterine artery pulsatility index (mUtPI) and biochemical markers including placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1) were measured at 11-13 + 6 weeks' gestation. Logistic regression analysis was performed to determine the significant predictors of complications. RESULTS Among 123 included pregnancies, 38 (30.9%) had GDM ± LGA fetus and 28 (22.8%) had GOS. The median maternal height and body mass index were significantly higher in women with GDM ± LGA fetus. Multivariate logistic regression analysis demonstrated that in the prediction of GDM ± LGA fetus and GOS, there were significant independent contributions from FI MoM (area under curve (AUROC) of 0.610, 95% CI 0.492-0.727; p = 0.062) and MAP MoM (AUROC of 0.645, 95% CI 0.510-0.779; p = 0.026), respectively. CONCLUSION FI and MAP are independent predictors for GDM ± LGA fetus and GOS, respectively. However, they have low predictive value. There is a need to identify more specific novel biomarkers in differentiating IVF/ET pregnancies that are at a higher risk of developing complications.
Collapse
Affiliation(s)
- Patricia Nga Ping Ip
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Long Nguyen-Hoang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Piya Chaemsaithong
- Department of Obstetrics and Gynecology, Mahidol University, Bangkok, Thailand
| | - Jun Guo
- Department of Obstetrics and Gynaecology, Beijing Tongren Hospital, The Capital Medical University, Beijing, China
| | - Xueqin Wang
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Daljit Singh Sahota
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jacqueline Pui Wah Chung
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Liona Chiu Yee Poon
- Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
27
|
Hong J, Crawford K, Cavanagh E, da Silva Costa F, Kumar S. Placental growth factor and fetoplacental Doppler indices in combination predict preterm birth reliably in pregnancies complicated by fetal growth restriction. ULTRASOUND IN OBSTETRICS & GYNECOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY OF ULTRASOUND IN OBSTETRICS AND GYNECOLOGY 2024; 63:635-643. [PMID: 37820083 DOI: 10.1002/uog.27513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/25/2023] [Accepted: 10/02/2023] [Indexed: 10/13/2023]
Abstract
OBJECTIVE To assess the association between placental biomarkers (placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFlt-1)/PlGF ratio) and fetoplacental Doppler indices (umbilical artery (UA) pulsatility index (PI) and uterine artery (UtA) PI) in various combinations for predicting preterm birth (PTB) in pregnancies complicated by fetal growth restriction (FGR). METHODS This was a prospective observational cohort study, performed at Mater Mother's Hospital in Brisbane, Queensland, Australia, from May 2022 to June 2023, of pregnancies complicated by FGR and appropriate-for-gestational-age (AGA) pregnancies. Maternal serum PlGF levels, sFlt-1/PlGF ratio, UA-PI and UtA-PI were measured at 2-4-weekly intervals from recruitment until delivery. Harrell's concordance statistic (Harrell's C) was used to evaluate multivariable Cox proportional hazards regression models featuring various combinations of placental biomarkers and fetoplacental Doppler indices to ascertain the best combination to predict PTB (< 37 weeks). Multivariable Cox regression models were used with biomarkers as time-varying covariates. RESULTS The study cohort included 320 singleton pregnancies, comprising 179 (55.9%) affected by FGR, defined according to a Delphi consensus, and 141 (44.1%) with an AGA fetus. In the FGR cohort, both low PlGF levels and elevated sFlt-1/PlGF ratio were associated with significantly shorter time to PTB. Low PlGF was a better predictor of PTB than was either sFlt-1/PlGF ratio or a combination of PlGF and sFlt-1/PlGF ratio (Harrell's C, 0.81, 0.78 and 0.79, respectively). Although both Doppler indices were significantly associated with time to PTB, in combination they were better predictors of PTB than was either UA-PI > 95th centile or UtA-PI > 95th centile alone (Harrell's C, 0.82, 0.75 and 0.76, respectively). Predictive utility for PTB was best when PlGF < 100 ng/L, UA-PI > 95th centile and UtA-PI > 95th centile were combined (Harrell's C, 0.88) (hazard ratio, 32.99; 95% CI, 10.74-101.32). CONCLUSIONS Low maternal serum PlGF level (< 100 ng/L) and abnormal fetoplacental Doppler indices (UA-PI > 95th centile and UtA-PI > 95th centile) in combination have the greatest predictive utility for PTB in pregnancies complicated by FGR. Their assessment may help guide clinical management of these complex pregnancies. © 2023 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.
Collapse
Affiliation(s)
- J Hong
- Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - K Crawford
- Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
| | - E Cavanagh
- Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
| | - F da Silva Costa
- School of Medicine and Dentistry, Griffith University and Maternal Fetal Medicine Unit, Gold Coast University Hospital, Gold Coast, Queensland, Australia
| | - S Kumar
- Mater Research Institute, University of Queensland, South Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Herston, Queensland, Australia
- NHMRC Centre for Research Excellence in Stillbirth, Mater Research Institute, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
28
|
Scher MS, Agarwal S, Venkatesen C. Clinical decisions in fetal-neonatal neurology I. reproductive and pregnancy health influence the neural exposome over multiple generations. Semin Fetal Neonatal Med 2024:101521. [PMID: 38658296 DOI: 10.1016/j.siny.2024.101521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Interdisciplinary fetal neonatal neurology (FNN) training requires integration of reproductive health factors into evaluations of the maternal-placental-fetal (MPF) triad, neonate, and child over the first 1000 days. Serial events that occur before one or multiple pregnancies impact successive generations. A maternal-child dyad history highlights this continuity of health risk, beginning with a maternal grandmother's pregnancy. Her daughter was born preterm and later experienced polycystic ovarian syndrome further complicated by cognitive and mental health disorders. Medical problems during her pregnancy contributed to MPF triad diseases that resulted in her son's extreme prematurity. Postpartum maternal death from the complications of diabetic ketoacidosis and her child's severe global neurodevelopmental delay were adverse mother-child outcomes. A horizontal/vertical diagnostic approach to reach shared clinical decisions during FNN training requires perspectives of a dynamic neural exposome. Career-long learning is then strengthened by continued interactions from al stakeholders. Developmental origins theory applied to neuroplasticity principles help interpret phenotypic expressions as dynamic gene-environment interactions across a person's lifetime. Debiasing strategies applied to the cognitive process reduce bias to preserve therapeutic and prognostic accuracy. Social determinants of health are essential components of this strategy to be initiated during FNN training. Reduction of the global burden of neurologic disorders requires applying the positive effects from reproductive and pregnancy exposomes that will benefit the neural exposome across the lifespan.
Collapse
Affiliation(s)
- Mark S Scher
- Rainbow Babies and Children's Hospital Case Western Reserve University School of Medicine, USA.
| | - Sonika Agarwal
- Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, USA.
| | - Charu Venkatesen
- Cincinnati Children's Hospital, Cincinnati School of Medicine, USA.
| |
Collapse
|
29
|
CHAIWORAPONGSA T, ROMERO R, GOMEZ-LOPEZ N, SUKSAI M, GALLO DM, JUNG E, BERRY SM, AWONUGA A, TARCA AL, BRYANT DR. Preeclampsia at term: evidence of disease heterogeneity based on the profile of circulating cytokines and angiogenic factors. Am J Obstet Gynecol 2024; 230:450.e1-450.e18. [PMID: 37806612 PMCID: PMC10990810 DOI: 10.1016/j.ajog.2023.10.002] [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: 08/08/2023] [Revised: 09/29/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Intravascular inflammation and an antiangiogenic state have been implicated in the pathophysiology of preeclampsia. On the basis of the profiles of their angiogenic/antiangiogenic factors, women with preeclampsia at term may be classified into 2 subgroups with different characteristics and prevalence of adverse outcomes. This study was undertaken to examine whether these 2 subgroups of preeclampsia at term also show differences in their profiles of intravascular inflammation. OBJECTIVE This study aimed to determine the plasma profiles of cytokines and chemokines in women with preeclampsia at term who had a normal or an abnormal angiogenic profile. STUDY DESIGN A nested case-control study was conducted to include women classified into 3 groups: women with an uncomplicated pregnancy (n=213) and women with preeclampsia at term with a normal (n=55) or an abnormal (n=41) angiogenic profile. An abnormal angiogenic profile was defined as a plasma ratio of placental growth factor and soluble fms-like tyrosine kinase-1 multiple of the median <10th percentile for gestational age. Concentrations of cytokines were measured by multiplex immunoassays. RESULTS Women with preeclampsia at term and an abnormal angiogenic profile showed evidence of the greatest intravascular inflammation among the study groups. These women had higher plasma concentrations of 5 cytokines (interleukin-6, interleukin-8, interleukin-12/interleukin-23p40, interleukin-15, and interleukin-16) and 7 chemokines (eotaxin, eotaxin-3, interferon-γ inducible protein-10, monocyte chemotactic protein-4, macrophage inflammatory protein-1β, macrophage-derived chemokine, and thymus and activation-regulated chemokine compared to women with an uncomplicated pregnancy. By contrast, women with preeclampsia at term and a normal angiogenic profile, compared to women with an uncomplicated pregnancy, had only a higher plasma concentration of monocyte chemotactic protein-4. A correlation between severity of the antiangiogenic state, blood pressure, and plasma concentrations of a subset of cytokines was observed. CONCLUSION Term preeclampsia can be classified into 2 clusters. One is characterized by an antiangiogenic state coupled with an excessive inflammatory process, whereas the other has neither of these features. These findings further support the heterogeneity of preeclampsia at term and may explain the distinct clinical outcomes.
Collapse
Affiliation(s)
- Tinnakorn CHAIWORAPONGSA
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto ROMERO
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
| | - Nardhy GOMEZ-LOPEZ
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
| | - Manaphat SUKSAI
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Dahiana M. GALLO
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Valle, Cali, Colombia
| | - Eunjung JUNG
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Stanley M. BERRY
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Awoniyi AWONUGA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. TARCA
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - David R. BRYANT
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
30
|
Chen JY, Yu BL, Wu XJ, Li YF, Zhong LY, Chen M. A longitudinal and cross-sectional study of placental circulation between normal and placental insufficiency pregnancies. Placenta 2024; 149:29-36. [PMID: 38490095 DOI: 10.1016/j.placenta.2024.03.001] [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: 09/05/2023] [Revised: 02/11/2024] [Accepted: 03/03/2024] [Indexed: 03/17/2024]
Abstract
INTRODUCTION To longitudinally and cross-sectionally study the differences in the uterine artery pulsatility index (UTPI), umbilical artery pulsatility index (UAPI) and placental vascularization indices (PVIs, derived from 3-dimensional power Doppler) between normal and placental insufficiency pregnancies throughout gestation. METHODS UTPI, UAPI and PVI were measured 6 times at 4- to 5- week intervals from 11 to 13+6 weeks-36 weeks. Preeclampsia (PE) and fetal growth restriction (FGR) were defined as placental insufficiency. Comparisons of UTPI, UAPI and PVI between normal and insufficiency groups were performed by one-way repeated measures analysis of variance. RESULTS A total of 125 women were included: monitored regularly from the first trimester to 36 weeks of gestation: 109 with normal pregnancies and 16 with placental insufficiency. Longitudinal study of the normal pregnancy group showed that UTPI and UAPI decreased significantly every 4 weeks, while PVIs increased significantly every 8 weeks until term. In the placental insufficiency group however, this decrease occurred slower at 8 weeks intervals and UTPI stabilized after 24 weeks. No significant difference was noted in PVIs throughout pregnancy. Cross-sectional study from different stages of gestation showed that UTPI was higher in the insufficiency group from 15 weeks onward and PVIs were lower after 32 weeks. DISCUSSION Compared to high-risk pregnancies with normal outcome, UTPI and UAPI needed a longer time to reach a significant change in those with clinical confirmation of placental insufficiency pregnancies and no significant change was found in PVI throughout gestation. UTPI was the earliest factor in detecting adverse outcome pregnancies.
Collapse
Affiliation(s)
- J Y Chen
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Guangzhou, China; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangzhou, China; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangzhou, China; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - B L Yu
- Department of Bio Resource Research Center, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - X J Wu
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Guangzhou, China; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangzhou, China; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangzhou, China; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Y F Li
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Guangzhou, China; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangzhou, China; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangzhou, China; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - L Y Zhong
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - M Chen
- Department of Obstetrics and Gynecology, Department of Fetal Medicine and Prenatal Diagnosis, Guangzhou, China; Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangzhou, China; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangzhou, China; Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, Guangzhou, China; The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
31
|
de Vos ES, van der Meeren LE, Koning AHJ, Nikkels PGJ, Steegers EAP, Steegers-Theunissen RPM, Mulders AGMGJ. First-trimester 3D power Doppler imaging markers of utero-placental vascular development are associated with placental weight and diameter at birth: The Rotterdam Periconception Cohort. Placenta 2024; 148:44-52. [PMID: 38367314 DOI: 10.1016/j.placenta.2024.01.019] [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: 09/18/2023] [Revised: 01/14/2024] [Accepted: 01/30/2024] [Indexed: 02/19/2024]
Abstract
INTRODUCTION Early utero-placental vascular development impacts placental development and function throughout pregnancy. We investigated whether impaired first-trimester utero-placental vascular development is associated with pathologic features of the postpartum placenta. METHODS In this prospective observational study of 65 ongoing pregnancies, we obtained three-dimensional power Doppler ultrasounds of the placenta at 7, 9 and 11 weeks of gestation. We applied VOCAL software to measure placental volume (PV), virtual reality based segmentation to measure utero-placental vascular volume (uPVV) and applied a skeletonization algorithm to generate the utero-placental vascular skeleton (uPVS). Vascular morphology was quantified by assigning a morphologic characteristic to each voxel in the uPVS (i.e. end-, bifurcation-, crossing- or vessel point). Following delivery, placentas were measured and histologically examined according to the Amsterdam criteria to assess maternal vascular malperfusion (MVM). We used linear mixed models to estimate trajectories of PV, uPVV and uPVS development. Multivariable linear regression analysis with adjustments for confounders was used to evaluate associations between PV, uPVV and uPVS development and features of the postpartum placenta. RESULTS We observed no associations between first-trimester PV development and measurements of the postpartum placenta. Increased first-trimester utero-placental vascular development, reflected by uPVV (β = 0.25 [0.01; 0.48]), uPVS end points (β = 0.25 [0.01; 0.48]), bifurcation points (β = 0.22 [0.05; 0.37]), crossing points (β = 0.29 [0.07; 0.52]) and vessel points (β = 0.09 [0.02; 0.17]) was positively associated with the postpartum placental diameter. uPVV was positively associated with postpartum placental weight. No associations were found with MVM. DISCUSSION Development of the first-trimester utero-placental vasculature is associated with postpartum placental size, whereas placental tissue development contributes to a lesser extent.
Collapse
Affiliation(s)
- Eline S de Vos
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, the Netherlands
| | - Lotte E van der Meeren
- Department of Pathology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, the Netherlands; Department of Pathology, Leiden University Medical Center, PO Box 9600, 2300 RC, Leiden, the Netherlands
| | - Anton H J Koning
- Department of Pathology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, the Netherlands
| | - Peter G J Nikkels
- Department of Pathology, University Medical Center Utrecht, H04.312, PO Box 85500, 3505 GA, Utrecht, the Netherlands
| | - Eric A P Steegers
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, the Netherlands
| | - Régine P M Steegers-Theunissen
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, the Netherlands
| | - Annemarie G M G J Mulders
- Department of Obstetrics and Gynecology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA, Rotterdam, the Netherlands.
| |
Collapse
|
32
|
Gallagher LT, Bardill J, Sucharov CC, Wright CJ, Karimpour-Fard A, Zarate M, Breckenfelder C, Liechty KW, Derderian SC. Dysregulation of miRNA-mRNA expression in fetal growth restriction in a caloric restricted mouse model. Sci Rep 2024; 14:5579. [PMID: 38448721 PMCID: PMC10918062 DOI: 10.1038/s41598-024-56155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/01/2024] [Indexed: 03/08/2024] Open
Abstract
Fetal growth restriction (FGR) is associated with aberrant placentation and accounts for a significant proportion of perinatal deaths. microRNAs have been shown to be dysregulated in FGR. The purpose of this study was to determine microRNA-regulated molecular pathways altered using a caloric restricted mouse model of FGR. Pregnant mice were subjected to a 50% caloric restricted diet beginning at E9. At E18.5, RNA sequencing of placental tissue was performed to identify differences in gene expression between caloric restricted and control placentas. Significant differences in gene expression between caloric restricted and control placentas were observed in 228 of the 1546 (14.7%) microRNAs. Functional analysis of microRNA-mRNA interactions demonstrated enrichment of several biological pathways with oxidative stress, apoptosis, and autophagy pathways upregulated and angiogenesis and signal transduction pathways downregulated. Ingenuity pathway analysis also suggested that ID1 signaling, a pathway integral for trophoblast differentiation, is also dysregulated in caloric restricted placentas. Thus, a maternal caloric restriction mouse model of FGR results in aberrant microRNA-regulated molecular pathways associated with angiogenesis, oxidative stress, signal transduction, apoptosis, and cell differentiation. As several of these pathways are dysregulated in human FGR, our findings suggest that this model may provide an excellent means to study placental microRNA derangements seen in FGR.
Collapse
Affiliation(s)
- Lauren T Gallagher
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - James Bardill
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Anis Karimpour-Fard
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Miguel Zarate
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Courtney Breckenfelder
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Kenneth W Liechty
- Division of Pediatric Surgery, University of Arizona College of Medicine, Tucson, AZ, 85721, USA
| | - S Christopher Derderian
- Colorado Fetal Care Center, Children's Hospital Colorado, University of Colorado, 13123 E 16th Ave, Aurora, CO, 80045, USA.
- Division of Pediatric Surgery, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA.
| |
Collapse
|
33
|
Hu J, Chen L, Ruan J, Chen X. The role of the annexin A protein family at the maternal-fetal interface. Front Endocrinol (Lausanne) 2024; 15:1314214. [PMID: 38495790 PMCID: PMC10940358 DOI: 10.3389/fendo.2024.1314214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 02/09/2024] [Indexed: 03/19/2024] Open
Abstract
Successful pregnancy requires the tolerance of the maternal immune system for the semi-allogeneic embryo, as well as a synchrony between the receptive endometrium and the competent embryo. The annexin family belongs to calcium-regulated phospholipid-binding protein, which functions as a membrane skeleton to stabilize the lipid bilayer and participate in various biological processes in humans. There is an abundance of the annexin family at the maternal-fetal interface, and it exerts a crucial role in embryo implantation and the subsequent development of the placenta. Altered expression of the annexin family and dysfunction of annexin proteins or polymorphisms of the ANXA gene are involved in a range of pregnancy complications. In this review, we summarize the current knowledge of the annexin A protein family at the maternal-fetal interface and its association with female reproductive disorders, suggesting the use of ANXA as the potential therapeutic target in the clinical diagnosis and treatment of pregnancy complications.
Collapse
Affiliation(s)
- Jingwen Hu
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
| | - Lin Chen
- Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jing Ruan
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
| | - Xiaoyan Chen
- Maternal-Fetal Medicine Institute, Department of Obstetrics and Gynaecology, Shenzhen Baoan Women’s and Children’s Hospital, Shenzhen University, Shenzhen, China
- Fertility Preservation Research Center, Department of Obstetrics and Gynaecology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
34
|
James AH, Strouse JJ. How I treat sickle cell disease in pregnancy. Blood 2024; 143:769-776. [PMID: 37979134 DOI: 10.1182/blood.2023020728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/15/2023] [Accepted: 10/12/2023] [Indexed: 11/19/2023] Open
Abstract
ABSTRACT Fifty years ago, people with sickle cell disease (SCD) were discouraged from becoming pregnant, but now, most should be supported if they choose to pursue a pregnancy. They and their providers, however, should be aware of the physiological changes of pregnancy that aggravate SCD and pregnancy's unique maternal and fetal challenges. Maternal problems can arise from chronic underlying organ dysfunction such as renal disease or pulmonary hypertension; from acute complications of SCD such as acute anemia, vaso-occlusive crises, and acute chest syndrome; and/or from pregnancy-related complications such as preeclampsia, sepsis, severe anemia, thromboembolism, and the need for cesarean delivery. Fetal problems include alloimmunization, opioid exposure, fetal growth restriction, preterm delivery, and stillbirth. Before and during pregnancy, in addition to the assessment and care that every pregnant patient should receive, patients with SCD should be evaluated and treated by a multidisciplinary team with respect to their unique maternal and fetal issues.
Collapse
Affiliation(s)
- Andra H James
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Duke University, Durham, NC
- Division of Hematology, Department of Medicine, Duke University, Durham, NC
| | - John Joseph Strouse
- Division of Hematology, Department of Medicine, Duke University, Durham, NC
- Division of Hematology/Oncology, Department of Pediatrics, Duke University, Durham, NC
| |
Collapse
|
35
|
Marsosi V, Haghighi L, Nasimi PH, Ghaemi M, Navaee S. Effects of sildenafil on Doppler parameters, maternal and neonatal outcomes in the active labor phase of low-risk pregnancies: a randomized clinical trial. J Perinat Med 2024; 52:210-214. [PMID: 37931599 DOI: 10.1515/jpm-2023-0210] [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: 05/28/2023] [Accepted: 09/12/2023] [Indexed: 11/08/2023]
Abstract
OBJECTIVES The benefits of sildenafil by increasing blood flow in the improvement of Doppler parameters of umbilical (UA), uterine (UtA), and fetal middle cerebral arteries (MCA) remain uncertain. On the other hand, insufficient blood flow during uterine contractions in labor can lead to decrease blood supply and fetal distress. Therefore, we aimed to assess the changes in fetal Doppler indices and maternal and neonatal outcomes following the use of sildenafil in the active phase of labor in low-risk pregnancies with healthy fetuses. METHODS This randomized double-blinded controlled trial was conducted on 70 pregnant single low-risk, pregnant women. The patients were randomly assigned into two groups receiving sildenafil (n=35) or placebo (n=35) when the active phase of labor was initiated. Doppler parameters were assessed at baseline as well as 3 h after that. Indeed, the maternal and neonatal outcomes were compared between groups. RESULTS The Doppler parameters including the pulsatility index of MCA, UA, and left and right UtA remained unchanged after the administration of sildenafil. Neonatal outcomes including birth weight, PH of the umbilical artery, Apgar score, respiratory distress syndrome, and neonatal intensive care unit admission as well as maternal outcomes such as cesarean section rate and the occurrence of intrapartum/postpartum hemorrhage had no difference between groups. CONCLUSIONS The use of sildenafil in the active phase of labor in low-risk pregnancies may not be beneficial in improving Doppler parameters in MCA, umbilical, and uterine arteries and thus may not improve pregnancy outcomes.
Collapse
Affiliation(s)
- Vajiheh Marsosi
- Department of Obstetrics and Gynecology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ladan Haghighi
- Shahid Akbarabadi Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Marjan Ghaemi
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Saimaz Navaee
- Department of Obstetrics and Gynecology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
de Ganzo Suárez T, de Paco Matallana C, Plasencia W. Spiral, uterine artery doppler and placental ultrasound in relation to preeclampsia. Best Pract Res Clin Obstet Gynaecol 2024; 92:102426. [PMID: 38039843 DOI: 10.1016/j.bpobgyn.2023.102426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/18/2023] [Accepted: 08/06/2023] [Indexed: 12/03/2023]
Abstract
Preeclampsia (PE) is a multiorgan disorder that complicates around 2-8% of pregnancies and is a major cause of perinatal and maternal morbidity and mortality. PE is a clinical syndrome characterized by hypertension secondary to systemic inflammation, endothelial dysfunction, and syncytiotrophoblast stress leading to hypertension and multiorgan dysfunction. The uterine arteries are the main blood vessels that supply blood to the uterus. They give off branches and plays an important role in maintaining blood supply during pregnancy. The arcuate artery originates from the uterine artery and runs medially through the myometrium. The arcuate arteries divide almost directly into anterior and posterior branches, from which the radial artery leads directly to the uterine cavity during their course. Near the endometrium-myometrium junction, the radial artery generates spiral arteries within the basal layer and functional endometrium. The walls of radial and spiral arteries are rich in smooth muscle, which is lost when trophoblast cells invade and become large-caliber vessels. This physiological transformation of uteroplacental spiral arteries is critical for successful placental implantation and normal placental function. In normal pregnancy, the luminal diameter of the spiral arteries is greatly increased, and the vascular smooth muscle is replaced by trophoblast cells. This process and changes in the spiral arteries are called spiral artery remodeling. In PE, this genetically and immunologically governed process is deficient and therefore there is decreased vascular capacitance and increased resistance in the uteroplacental circulation. Furthermore, this defect in uteroplacental spiral artery remodeling is not only associated with early onset PE, but also with fetal growth restriction, placental abruption, and spontaneous premature rupture of membranes. Doppler ultrasound allows non-invasive assessment of placentation, while the flow impedance decreases as the pregnancy progresses in normal pregnancies, in those destined to develop preeclampsia the impedance is increased.
Collapse
Affiliation(s)
- Tania de Ganzo Suárez
- Department of Obstetrics and Gynecology, Complejo Hospitalario Universitario Nuestra Señora de la Candelaria, Tenerife, Canary Islands, Spain.
| | - Catalina de Paco Matallana
- Department of Obstetrics and Gynecology, Hospital Clínico Universitario Virgen de la Arrixaca, El Palmar, Murcia, Spain; Institute for Biomedical Research of Murcia, IMIB-Arrixaca, El Palmar, Murcia, Spain; Faculty of Medicine, Universidad de Murcia, Murcia, Spain.
| | - Walter Plasencia
- Department of Obstetrics and Gynecology, Complejo Hospitalario Universitario de Canarias, Tenerife. Canary Islands, Spain.
| |
Collapse
|
37
|
Scher MS, Agarwal S, Venkatesen C. Clinical decisions in fetal-neonatal neurology II: Gene-environment expression over the first 1000 days presenting as "four great neurological syndromes". Semin Fetal Neonatal Med 2024; 29:101522. [PMID: 38637242 DOI: 10.1016/j.siny.2024.101522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Interdisciplinary fetal-neonatal neurology (FNN) training considers a woman's reproductive and pregnancy health histories when assessing the "four great neonatal neurological syndromes". This maternal-child dyad exemplifies the symptomatic neonatal minority, compared with the silent majority of healthy children who experience preclinical diseases with variable expressions over the first 1000 days. Healthy maternal reports with reassuring fetal surveillance testing preceded signs of fetal distress during parturition. An encephalopathic neonate with seizures later exhibited childhood autistic spectrum behaviors and intractable epilepsy correlated with identified genetic biomarkers. A systems biology approach to etiopathogenesis guides the diagnostic process to interpret phenotypic form and function. Evolving gene-environment interactions expressed by changing phenotypes reflect a dynamic neural exposome influenced by reproductive and pregnancy health. This strategy considers critical/sensitive periods of neuroplasticity beyond two years of life to encompass childhood and adolescence. Career-long FNN experiences reenforce earlier training to strengthen the cognitive process and minimize cognitive biases when assessing children or adults. Prioritizing social determinants of healthcare for persons with neurologic disorders will help mitigate the global burden of brain diseases for all women and children.
Collapse
Affiliation(s)
- Mark S Scher
- Pediatrics and Neurology, Rainbow Babies and Children's Hospital Case Western Reserve University School of Medicine, USA.
| | - Sonika Agarwal
- Neurology and Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, USA.
| | - Charu Venkatesen
- Neurology and Pediatrics, Cincinnati Children's Hospital, Cincinnati School of Medicine, USA.
| |
Collapse
|
38
|
Huang S, Cai S, Ling L, Zhang W, Xiao H, Yu D, Zhong X, Tao P, Luo Y. Investigating the molecular mechanism of traditional Chinese medicine for the treatment of placental syndromes by influencing inflammatory cytokines using the Mendelian randomization and molecular docking technology. Front Endocrinol (Lausanne) 2024; 14:1290766. [PMID: 38362587 PMCID: PMC10868387 DOI: 10.3389/fendo.2023.1290766] [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: 09/08/2023] [Accepted: 12/29/2023] [Indexed: 02/17/2024] Open
Abstract
Introduction Placental syndromes, which include pregnancy loss, preterm birth, gestational diabetes mellitus (GDM), and hypertensive disorders in pregnancy (HDP), have a strong association with disorder inflammatory reactions. Nonetheless, the exact causal relationship has not been established. This study aims to investigate the causal relationship between placental syndromes and inflammatory cytokines utilizing Mendelian randomization (MR). Additionally, we examined the interaction between small molecular compounds derived from traditional Chinese medicine and inflammatory cytokines using molecular docking method. Methods After obtaining the data of inflammatory cytokines and placental syndromes, as well as establishing single nucleotide polymorphisms (SNPs), we employed the inverse variance weighted (IVW) method to assess the causal relationship. We also accessed the heterogeneity and the horizontal pleiotropy of these data. The "ClusterProfiler" R package was utilized for Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) term analyses. The protein-protein interaction (PPI) network was constructed using STRING database. AutoDock Vina software was used for molecular docking, and Discovery Studio 2019 was used for visualization purposes. Results We found that the growth regulated oncogene A (GROA) and interleukin-9 (IL-9) were associated with the development of pregnancy hypertension, whereas interleukin-10 (IL-10) and hepatocyte growth factor (HGF) were linked to the occurrence of preeclampsia. Moreover, there were correlations observed between interleukin-18 (IL-18), IL-10, macrophage colony-stimulating factor (MCSF), and platelet-derived growth factor BB (PDGFbb) in cases of chronic hypertension combined with pregnancy (CHP). Additionally, macrophage migration inhibitory factor (MIF) exhibited a connection with GDM, and TNF related apoptosis inducing ligand (TRAIL) demonstrated a causal relationship with preterm birth. It is plausible to suggest that interleukin-1β (IL-1β) might contribute to the promotion of pregnancy loss. All of the binding free energy values of small molecular compounds with inflammatory cytokines were below -5.0 kcal/mol. Furthermore, all of the RMSD values were less than 2. Conclusions GROA, IL-1β, IL-9, IL-10, IL-18, MIF, MCSF, HGF, PDGFbb and TRAIL were found to be causally associated with placental syndromes. Molecular docking analysis revealed that small molecular compounds, such as puerarin, magnolol, atractylenolide I, paeoniflorin, tumulosic acid and wogonin, are closely bound to these inflammatory cytokines.
Collapse
Affiliation(s)
- Shan Huang
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Shuangming Cai
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Lin Ling
- Department of Rehabilitation, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenni Zhang
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Huanshun Xiao
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Danfeng Yu
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Xuan Zhong
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Pei Tao
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Yiping Luo
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
39
|
James AH, James PD. What do we know about why women bleed and what do we not know? J Thromb Haemost 2024; 22:315-322. [PMID: 37709147 DOI: 10.1016/j.jtha.2023.08.034] [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: 06/08/2023] [Revised: 08/07/2023] [Accepted: 08/31/2023] [Indexed: 09/16/2023]
Abstract
Women or people with a uterus are vulnerable to both normal and abnormal bleeding. During the reproductive years, the uterus is prepared physiologically to accept an embryo and support its growth and development during pregnancy, or in the absence of implantation of an embryo, recycle through the process of menstruation and accept an embryo a month or so later. If fertilization takes place and an embryo or embryos implant in the uterus, the fetal trophoblast, or outer cell layer of the embryo, invades and dilates the maternal spiral arteries and forms the placenta. No matter when in gestation a pregnancy ends, at the conclusion of pregnancy, the placenta should separate from the wall of the uterus and be expelled. Abnormal bleeding occurs during pregnancy or after delivery when the normal uteroplacental interface has not been established or is interrupted; during miscarriage; during ectopic pregnancy; during premature separation of the placenta; or during postpartum hemorrhage. Heavy menstrual bleeding, a subset of abnormal menstrual bleeding, can be quantitatively defined as >80 mL of blood loss per cycle. Unlike postpartum hemorrhage, heavy menstrual bleeding is significantly associated with an underlying bleeding disorder. While there is other reproductive tract bleeding in women, notably bleeding at the time of ovulation or with a life-threatening ruptured ectopic pregnancy, the unique bleeding that women experience is predominantly uterine in origin. Many of the unique aspects of uterine hemostasis, however, remain unknown.
Collapse
Affiliation(s)
- Andra H James
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, Durham, USA; Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, Durham, USA.
| | - Paula D James
- Departments of Medicine and Pathology & Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
40
|
Scher MS. The science of uncertainty guides fetal-neonatal neurology principles and practice: diagnostic-prognostic opportunities and challenges. Front Neurol 2024; 15:1335933. [PMID: 38352135 PMCID: PMC10861710 DOI: 10.3389/fneur.2024.1335933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/08/2024] [Indexed: 02/16/2024] Open
Abstract
Fetal-neonatal neurologists (FNNs) consider diagnostic, therapeutic, and prognostic decisions strengthened by interdisciplinary collaborations. Bio-social perspectives of the woman's health influence evaluations of maternal-placental-fetal (MPF) triad, neonate, and child. A dual cognitive process integrates "fast thinking-slow thinking" to reach shared decisions that minimize bias and maintain trust. Assessing the science of uncertainty with uncertainties in science improves diagnostic choices across the developmental-aging continuum. Three case vignettes highlight challenges that illustrate this approach. The first maternal-fetal dyad involved a woman who had been recommended to terminate her pregnancy based on an incorrect diagnosis of an encephalocele. A meningocele was subsequently identified when she sought a second opinion with normal outcome for her child. The second vignette involved two pregnancies during which fetal cardiac rhabdomyoma was identified, suggesting tuberous sclerosis complex (TSC). One woman sought an out-of-state termination without confirmation using fetal brain MRI or postmortem examination. The second woman requested pregnancy care with postnatal evaluations. Her adult child experiences challenges associated with TSC sequelae. The third vignette involved a prenatal diagnosis of an open neural tube defect with arthrogryposis multiplex congenita. The family requested prenatal surgical closure of the defect at another institution at their personal expense despite receiving a grave prognosis. The subsequent Management of Myelomeningocele Study (MOMS) would not have recommended this procedure. Their adult child requires medical care for global developmental delay, intractable epilepsy, and autism. These three evaluations involved uncertainties requiring shared clinical decisions among all stakeholders. Falsely negative or misleading positive interpretation of results reduced chances for optimal outcomes. FNN diagnostic skills require an understanding of dynamic gene-environment interactions affecting reproductive followed by pregnancy exposomes that influence the MPF triad health with fetal neuroplasticity consequences. Toxic stressor interplay can impair the neural exposome, expressed as anomalous and/or destructive fetal brain lesions. Functional improvements or permanent sequelae may be expressed across the lifespan. Equitable and compassionate healthcare for women and families require shared decisions that preserve pregnancy health, guided by person-specific racial-ethnic, religious, and bio-social perspectives. Applying developmental origins theory to neurologic principles and practice supports a brain health capital strategy for all persons across each generation.
Collapse
Affiliation(s)
- Mark Steven Scher
- Fetal/Neonatal Neurology Program, Division of Pediatric Neurology, Department of Pediatrics, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
41
|
Lan X, Guo L, Hu C, Zhang Q, Deng J, Wang Y, Chen ZJ, Yan J, Li Y. Fibronectin mediates activin A-promoted human trophoblast migration and acquisition of endothelial-like phenotype. Cell Commun Signal 2024; 22:61. [PMID: 38263146 PMCID: PMC10807102 DOI: 10.1186/s12964-023-01463-z] [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: 09/28/2023] [Accepted: 12/27/2023] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND During human early placentation, a proportion of extravillous trophoblasts (EVTs) migrate to the maternal decidua, differentiating into endovascular EVTs to remodel spiral arteries and ensure the establishment of blood circulation at the maternal-fetal interface. Inadequate EVT migration and endovascular differentiation are closely associated with adverse pregnancy outcomes such as miscarriage. Activin A and fibronectin are both secretory molecules abundantly expressed at the maternal-fetal interface. Activin A has been reported to regulate EVT biological functions. However, whether fibronectin mediates activin A-promoted EVT migration and acquisition of endothelial-like phenotype as well as the underlying molecular mechanisms remain unknown. Additionally, the role of fibronectin in pregnancy establishment and maintenance warrants further investigation. METHODS Primary and immortalized (HTR8/SVneo) human EVTs were used as in vitro study models. Cultured human first-trimester chorionic villous explants were utilized for ex vivo validation. A local fibronectin knockdown model in ICR mouse uteri, achieved by nonviral in vivo transfection with small interfering RNA (siRNA) targeting fibronectin 1 (si-Fn1), was employed to explore the roles of fibronectin in the establishment and maintenance of early pregnancy. RESULTS Our results showed that activin A treatment significantly induced fibronectin 1 (FN1) mRNA expression and fibronectin protein production, which is essential for human trophoblast migration and endothelial-like tube formation. Both basal and activin A-upregulated fibronectin expression were abolished by the TGF-β type I receptor inhibitor SB431542 or siRNA-mediated knockdown of activin receptor-like kinase (ALK4) or SMAD4. Moreover, activin A-increased trophoblast migration and endothelial-like tube formation were attenuated following the depletion of fibronectin. Fibronectin knockdown via intrauterine siRNA administration reduced CD31 and cytokeratin 8 (CK8) expression at the maternal-fetal interface, resulting in a decrease in the number of implantation sites and embryos. CONCLUSIONS Our study demonstrates that activin A promotes trophoblast cell migration and acquisition of endothelial-like phenotype via ALK4-SMAD2/3-SMAD4-mediated fibronectin upregulation. Furthermore, through a local fibronectin knockdown model in mouse uteri, we found that the absence of fibronectin at the maternal-fetal interface impedes endovascular migration of trophoblasts and decidual vascularization, thereby interfering with early embryo implantation and the maintenance of pregnancy. These findings provide novel insights into placental development during early pregnancy establishment and contribute to the advancement of therapeutic approaches for managing pregnancy complications related to trophoblast dysfunction.
Collapse
Affiliation(s)
- Xiangxin Lan
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Ling Guo
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Cuiping Hu
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Qian Zhang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Jianye Deng
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Yufeng Wang
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
| | - Zi-Jiang Chen
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, 250012, Shandong, China
| | - Junhao Yan
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China.
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China.
| | - Yan Li
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, Shandong, China.
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, 250012, Shandong, China.
- Medical Integration and Practice Center, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
42
|
Zhang W, Liu Z, Wang B, Liu M, Li J, Guan Y. Comparison of the perinatal outcomes of expected high ovarian response patients and normal ovarian response patients undergoing frozen-thawed embryo transfer in natural/small amount of HMG induced ovulation cycles. BMC Public Health 2024; 24:259. [PMID: 38254007 PMCID: PMC10804831 DOI: 10.1186/s12889-024-17725-5] [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: 09/05/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Due to the high risk of complications in fresh transfer cycles among expected high ovarian response patients, most choose frozen-thawed embryo transfer (FET). There are currently few researches on whether the FET outcomes of expected high ovarian response patients with regular menstrual cycles are similar to those of normal ovarian response. Therefore, our objective was to explore and compare pregnancy outcomes and maternal and neonatal outcomes of natural FET cycles between patients with expected high ovarian response and normal ovarian response with regular menstrual cycles based on the antral follicle count (AFC). METHODS This retrospective cohort study included 5082 women undergoing natural or small amount of HMG induced ovulation FET cycles at the Reproductive Center of the Third Affiliated Hospital of Zhengzhou University from January 1, 2017, to March 31, 2021. The population was divided into expected high ovarian response group and normal ovarian response group based on the AFC, and the differences in patient characteristics, clinical outcomes and perinatal outcomes between the two groups were compared. RESULTS Regarding clinical outcomes, compared with the normal ovarian response group, patients in the expected high ovarian response group had a higher clinical pregnancy rate (57.34% vs. 48.50%) and live birth rate (48.12% vs. 38.97%). There was no difference in the early miscarriage rate or twin pregnancy rate between the groups. Multivariate logistic regression analysis suggested that the clinical pregnancy rate (adjusted OR 1.190) and live birth rate (adjusted OR 1.171) of the expected high ovarian response group were higher than those of the normal ovarian response group. In terms of maternal and infant outcomes, the incidence of very preterm delivery in the normal ovarian response group was higher than that in the expected high ovarian response group (0.86% vs. 0.16%, adjusted OR 0.131), Other maternal and infant outcomes were not significantly different. After grouping by age (< 30 y, 30-34 y, 35-39 y), there was no difference in the incidence of very preterm delivery among the age subgroups. CONCLUSION For patients with expected high ovarian response and regular menstrual cycles undergoing natural or small amount of HMG induced ovulation FET cycles, the clinical and perinatal outcomes are reassuring. For patients undergoing natural or small amount of HMG induced ovulation FET cycles, as age increases, perinatal care should be strengthened during pregnancy to reduce the incidence of very preterm delivery.
Collapse
Affiliation(s)
- Wenjuan Zhang
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, Henan, China
| | - Zhaozhao Liu
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, Henan, China
| | - Bijun Wang
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, Henan, China
| | - Manman Liu
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, Henan, China
| | - Jiaheng Li
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, Henan, China
| | - Yichun Guan
- Reproduction Center, The Third Affiliated Hospital of ZhengZhou University, Henan, China.
| |
Collapse
|
43
|
Scher MS. Interdisciplinary fetal-neonatal neurology training applies neural exposome perspectives to neurology principles and practice. Front Neurol 2024; 14:1321674. [PMID: 38288328 PMCID: PMC10824035 DOI: 10.3389/fneur.2023.1321674] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 12/07/2023] [Indexed: 01/31/2024] Open
Abstract
An interdisciplinary fetal-neonatal neurology (FNN) program over the first 1,000 days teaches perspectives of the neural exposome that are applicable across the life span. This curriculum strengthens neonatal neurocritical care, pediatric, and adult neurology training objectives. Teaching at maternal-pediatric hospital centers optimally merges reproductive, pregnancy, and pediatric approaches to healthcare. Phenotype-genotype expressions of health or disease pathways represent a dynamic neural exposome over developmental time. The science of uncertainty applied to FNN training re-enforces the importance of shared clinical decisions that minimize bias and reduce cognitive errors. Trainees select mentoring committee participants that will maximize their learning experiences. Standardized questions and oral presentations monitor educational progress. Master or doctoral defense preparation and competitive research funding can be goals for specific individuals. FNN principles applied to practice offer an understanding of gene-environment interactions that recognizes the effects of reproductive health on the maternal-placental-fetal triad, neonate, child, and adult. Pre-conception and prenatal adversities potentially diminish life-course brain health. Endogenous and exogenous toxic stressor interplay (TSI) alters the neural exposome through maladaptive developmental neuroplasticity. Developmental disorders and epilepsy are primarily expressed during the first 1,000 days. Communicable and noncommunicable illnesses continue to interact with the neural exposome to express diverse neurologic disorders across the lifespan, particularly during the critical/sensitive time periods of adolescence and reproductive senescence. Anomalous or destructive fetal neuropathologic lesions change clinical expressions across this developmental-aging continuum. An integrated understanding of reproductive, pregnancy, placental, neonatal, childhood, and adult exposome effects offers a life-course perspective of the neural exposome. Exosome research promises improved disease monitoring and drug delivery starting during pregnancy. Developmental origins of health and disease principles applied to FNN practice anticipate neurologic diagnoses with interventions that can benefit successive generations. Addressing health care disparities in the Global South and high-income country medical deserts require constructive dialogue among stakeholders to achieve medical equity. Population health policies require a brain capital strategy that reduces the global burden of neurologic diseases by applying FNN principles and practice. This integrative neurologic care approach will prolong survival with an improved quality of life for persons across the lifespan confronted with neurological disorders.
Collapse
Affiliation(s)
- Mark S. Scher
- Division of Pediatric Neurology, Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
44
|
Jing Jia, Ma B, Zhao X. Fetal endothelial colony-forming cells: Possible targets for prevention of the fetal origins of adult diseases. Placenta 2024; 145:80-88. [PMID: 38100962 DOI: 10.1016/j.placenta.2023.12.006] [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: 08/10/2023] [Revised: 11/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
Endothelial colony-forming cells (ECFCs), a subset of circulating and resident endothelial progenitor cells, are capable of self-renewal and de novo vessel formation, and are known key regulators of vascular integrity and homeostasis. Numerous studies have found that exposure to hostile environment during the fetal development exerts a profound influence on the level and function of ECFCs, which may be the underlying factor linking endothelial dysfunction to cardiovascular disease of the offspring in later life. Herein, we focus on the latest findings regarding the effects of pregnancy-related disorders on the frequency and function of fetal ECFCs. Subsequently, we discuss about placental ECFCs and put forward some details that should be paid attention to in the process of ECFC isolation and culture. Overall, the information presented in this review highlight the potential of ECFCs as a future biomarker or even therapeutic targets for the pregnancy-related adverse maternal and fetal outcomes.
Collapse
Affiliation(s)
- Jing Jia
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Baitao Ma
- Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianlan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
45
|
Chiorean DM, Cobankent Aytekin E, Mitranovici MI, Turdean SG, Moharer MS, Cotoi OS, Toru HS. Human Placenta and Evolving Insights into Pathological Changes of Preeclampsia: A Comprehensive Review of the Last Decade. Fetal Pediatr Pathol 2024; 43:33-46. [PMID: 37906285 DOI: 10.1080/15513815.2023.2274823] [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: 09/07/2023] [Accepted: 10/18/2023] [Indexed: 11/02/2023]
Abstract
The placenta, the foremost and multifaceted organ in fetal and maternal biology, is pivotal in facilitating optimal intrauterine fetal development. Remarkably, despite its paramount significance, the placenta remains enigmatic, meriting greater comprehension given its central influence on the health trajectories of both the fetus and the mother. Preeclampsia (PE) and intrauterine fetal growth restriction (IUGR), prevailing disorders of pregnancy, stem from compromised placental development. PE, characterized by heightened mortality and morbidity risks, afflicts 5-7% of global pregnancies, its etiology shrouded in ambiguity. Pertinent pathogenic hallmarks of PE encompass inadequate restructuring of uteroplacental spiral arteries, placental ischemia, and elevated levels of vascular endothelial growth factor receptor-1 (VEGFR-1), also recognized as soluble FMS-like tyrosine kinase-1 (sFlt-1). During gestation, the placental derivation of sFlt-1 accentuates its role as an inhibitory receptor binding to VEGF-A and placental growth factor (PlGF), curtailing target cell accessibility. This review expounds upon the placenta's defining cellular component of the trophoblast, elucidates the intricacies of PE pathogenesis, underscores the pivotal contribution of sFlt-1 to maternal pathology and fetal safeguarding, and surveys recent therapeutic strides witnessed in the past decade.
Collapse
Affiliation(s)
- Diana Maria Chiorean
- Department of Pathology, County Clinical Hospital of Targu Mures, Targu Mures, Romania
| | | | | | - Sabin Gligore Turdean
- Department of Pathology, County Clinical Hospital of Targu Mures, Targu Mures, Romania
| | | | - Ovidiu Simion Cotoi
- Department of Pathology, County Clinical Hospital of Targu Mures, Targu Mures, Romania
- Department Pathophysiology, "George Emil Palade" University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania, and
| | - Havva Serap Toru
- Department of Pathology, School of Medicine, Akdeniz University, Antalya Pınarbaşı, Konyaaltı/Antalya, Turkey
| |
Collapse
|
46
|
Youssim I, Erez O, Novack L, Nevo D, Kloog I, Raz R. Ambient temperature and preeclampsia: A historical cohort study. ENVIRONMENTAL RESEARCH 2023; 238:117107. [PMID: 37696321 DOI: 10.1016/j.envres.2023.117107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 08/21/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
Previous studies found inconsistent associations between ambient temperature during pregnancy and the risk of preeclampsia. If such associations are causal, they may impact the future burden of preeclampsia in the context of climate change. We used a historical cohort of 129,009 pregnancies (5074 preeclampsia cases) from southern Israel that was merged with temperature assessments from a hybrid satellite-based exposure model. Distributed-lag and cause-specific hazard models were employed to study time to all preeclampsia cases, followed by stratification according to early (≤34 weeks) and late (>34 weeks) onset disease and identify critical exposure periods. We found a positive association between temperature and preeclampsia during gestation, which was stronger in the 3rd trimester. For example, during week 33, compared to the reference temperature of 22.4 °C, the cause-specific hazard ratio (HRCS) of preeclampsia was 1.01 (95% confidence interval (CI): 1.01-1.02) when exposed to 30 °C, 1.05 (95%CI: 1.03-1.08) at 35 °C, and 1.07 (95%CI: 1.04-1.10) at 37 °C. The associations existed with both early- and late-onset preeclampsia; however, the associations with the early-onset disease were somewhat stronger, limited to the first weeks of pregnancy and the third trimester, and with larger confidence intervals. The HRCS for early preeclampsia onset, when exposed to 37 °C compared to 22.4 °C during week 33, was 1.12 (95%CI: 0.96-1.30), and for late-onset preeclampsia, the HRCS was 1.09 (95%CI: 1.05-1.13). To conclude, exposure to high temperatures at the beginning and, particularly, the end of gestation is associated with an increased risk of preeclampsia in southern Israel.
Collapse
Affiliation(s)
- Iaroslav Youssim
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Israel.
| | - Offer Erez
- Division of Obstetrics and Gynecology, Soroka University Medical Center, Israel; Department of Obstetrics and Gynecology, Hutzel Women's Hospital, Wayne State University, Detroit, MI, USA
| | - Lena Novack
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Israel; Soroka University Medical Center, Israel, Ben-Gurion University of the Negev, Israel
| | - Daniel Nevo
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv, Israel
| | - Itai Kloog
- The Department of Geography and Environmental Development, Ben-Gurion University of the Negev, Israel
| | - Raanan Raz
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Israel
| |
Collapse
|
47
|
Baxter C, Crary I, Coler B, Marcell L, Huebner EM, Rutz S, Adams Waldorf KM. Addressing a broken drug pipeline for preterm birth: why early preterm birth is an orphan disease. Am J Obstet Gynecol 2023; 229:647-655. [PMID: 37516401 PMCID: PMC10818026 DOI: 10.1016/j.ajog.2023.07.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/19/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
Preterm birth remains one of the most urgent unresolved medical problems in obstetrics, yet only 2 therapeutics for preventing preterm birth have ever been approved by the United States Food and Drug Administration, and neither remains on the market. The recent withdrawal of 17-hydroxyprogesterone caproate (17-OHPC, Makena) marks a new but familiar era for obstetrics with no Food and Drug Administration-approved pharmaceuticals to address preterm birth. The lack of pharmaceuticals reflects a broad and ineffective pipeline hindered by extensive regulatory hurdles, soaring costs of performing drug research, and concerns regarding adverse effects among a particularly vulnerable population. The pharmaceutical industry has historically limited investments in research for diseases with similarly small markets, such as cystic fibrosis, given their rarity and diminished projected financial return. The Orphan Drug Act, however, incentivizes drug development for "orphan diseases", defined as affecting <200,000 people in the United States annually. Although the total number of preterm births in the United States exceeds this threshold annually, the early subset of preterm birth (<34 weeks' gestation) would qualify, which is predominantly caused by inflammation and infection. The scientific rationale for classifying preterm birth into early and late subsets is strong given that their etiologies differ, and therapeutics that may be efficacious for one subset may not work for the other. For example, antiinflammatory therapeutics would be expected to be highly effective for early but not late preterm birth. A robust therapeutic pipeline of antiinflammatory drugs already exists, which could be used to target spontaneous early preterm birth, in combination with antibiotics shown to sterilize the amniotic cavity. New applications for therapeutics targeting spontaneous early preterm birth could categorize as orphan disease drugs, which could revitalize the preterm birth therapeutic pipeline. Herein, we describe why drugs targeting early preterm birth should qualify for orphan status, which may increase pharmaceutical interest for this vitally important obstetrical condition.
Collapse
Affiliation(s)
- Carly Baxter
- School of Medicine, University of Washington, Seattle, WA
| | - Isabelle Crary
- School of Medicine, University of Washington, Seattle, WA
| | - Brahm Coler
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA
| | - Lauren Marcell
- School of Medicine, University of Washington, Seattle, WA
| | | | - Sara Rutz
- School of Medicine, University of Washington, Seattle, WA
| | - Kristina M Adams Waldorf
- Departments of Obstetrics and Gynecology and Global Health, University of Washington, Seattle, WA.
| |
Collapse
|
48
|
Turan OM, Babischkin JS, Aberdeen GW, Turan S, Pepe GJ, Albrecht ED. B-Flow/Spatiotemporal Image Correlation M-Mode and Contrast-Enhanced/Microbubble Ultrasonography Quantification of Spiral Artery Distensibility and Placental Intervillous Perfusion in the First Trimester in a Primate Model of Impaired Spiral Artery Remodeling. ULTRASOUND IN MEDICINE & BIOLOGY 2023; 49:2557-2564. [PMID: 37749012 PMCID: PMC10591761 DOI: 10.1016/j.ultrasmedbio.2023.08.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 08/17/2023] [Accepted: 08/28/2023] [Indexed: 09/27/2023]
Abstract
OBJECTIVE During early human pregnancy, placental trophoblasts remodel spiral arteries into distensible low-resistance vessels to promote placental perfusion. We have established a model of impaired spiral artery remodeling (SAR) by elevating estradiol levels in the first trimester of baboon pregnancy. In the present study, B-flow/spatiotemporal image correlation (STIC) M-mode ultrasonography, a non-Doppler technology for sharp rendering of vessel dimensions, was used to determine whether spiral artery distensibility was altered in SAR-suppressed baboons. Contrast-enhanced ultrasound/microbubble imaging was also performed to determine whether it detected changes in placenta intervillous space perfusion in SAR-suppressed baboons. METHODS The two imaging procedures were performed in the first trimester in baboons not treated or treated with estradiol to suppress SAR. RESULTS Spiral artery distensibility, that is, luminal diameter at systole minus diameter at diastole, and volume flow as quantified by B-flow/STIC M-mode were 26% (p = 0.03) and 55% (p = 0.059) lower, respectively, in SAR-suppressed baboons. However, placental intervillous space flow rate and video intensity plateau levels reflecting blood perfusion, quantified by contrast-enhanced ultrasound/microbubble imaging, were unaltered in SAR-suppressed baboons. CONCLUSION The results indicate that B-flow/STIC M-mode ultrasonography provides a non-invasive method to detect reduced distensibility and, thus, function of spiral arteries across the cardiac cycle in the first trimester in a primate model of impaired SAR. This study represents a first step in determining whether B-flow/STIC M-mode detects a similar defect in SAR early in adverse human pregnancy. This would provide an avenue to develop therapeutic modalities to prevent the devastating consequences of impaired SAR.
Collapse
Affiliation(s)
- Ozhan M Turan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeffery S Babischkin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Graham W Aberdeen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sifa Turan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gerald J Pepe
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Eugene D Albrecht
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
49
|
Álvarez SC, Zurita AR, Del Carmen De Ganzo Suárez T, De Luis Escudero JF, Medina NS, Pérez CC, de Basoa CMF, Montesino JLT, Masip MTC, Bello MÁG. Is a sFlt-1/PlGF cutoff of 38 suitable to predict adverse outcomes in pregnancies with abnormal uterine artery Doppler velocimetry in the second trimester? Pregnancy Hypertens 2023; 34:13-18. [PMID: 37778280 DOI: 10.1016/j.preghy.2023.09.006] [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: 07/27/2022] [Revised: 07/03/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVE To determine the optimal cutoff value for the soluble fms-like tyrosine kinase-1 (sFlt-1)/placental growth factor (PlGF) ratio to predict maternal and fetal adverse events in pregnancies with uterine artery Doppler scans results above the 95th percentile in the late second trimester. STUDY DESIGN Retrospective, observational cohort study on 116 asyntomatic patients with abnormal uterine artery Doppler scans at gestational week 25. The sFlt-1/PlGF ratio was determined within the weeks 25 to 29 of gestation and ROC curve analysis performed. The diagnostic validity of different cutoff values to predict severe maternal and fetal complications, i.e. preeclampsia, fetal growth restriction, placental abruption, and fetal death, was analyzed. MAIN OUTCOME MEASURES An ideal cutoff for sFlt-1/PlGF ratios in pregnancies with abnormal uterine artery Doppler in the second trimester. RESULTS Applying a cutoff point of 38, the area under the ROC curve was 0.89, generally considered low risk in fetal and maternal complication prediction. The sensitivity was 32.1%, the specificity 98.4%, the positive predictive value (PPV) 94.4%, and the negative predictive value (NPV) 63.3%. A cutoff value of 10, leading to the highest Youden index, performed best at detecting overall complications, increasing sensitivity to 69.8% and the NPV to 76.8%. at the cost of a reduced specificity and PPV. CONCLUSIONS In pregnancies with abnormal uterine artery Doppler in the second trimester, an sFlt-1/PlGF cutoff value greater than equal to 38 improves its predictive power for adverse events.
Collapse
Affiliation(s)
- Sara Caamiña Álvarez
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.
| | - Alicia Rodríguez Zurita
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | | | - José Fernando De Luis Escudero
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Nieves Sierra Medina
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Carolina Chulilla Pérez
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Cecilia Martín Fernández de Basoa
- Prenatal Screening and Preeclampsia Unit, Clinical Analysis Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - José Luis Trabado Montesino
- Department of Obstetrics and Gynecology, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - María Teresa Concepción Masip
- Prenatal Screening and Preeclampsia Unit, Clinical Analysis Service, University Hospital Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Miguel Ángel García Bello
- Department of Clinical Psychology-Psychobiology and Methodology, University of La Laguna, Santa Cruz de Tenerife, Spain
| |
Collapse
|
50
|
Juusela A, Jung E, Gallo DM, Bosco M, Suksai M, Diaz-Primera R, Tarca AL, Than NG, Gotsch F, Romero R, Chaiworapongsa T. Maternal plasma syndecan-1: a biomarker for fetal growth restriction. J Matern Fetal Neonatal Med 2023; 36:2150074. [PMID: 36597808 DOI: 10.1080/14767058.2022.2150074] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The identification of fetal growth disorders is an important clinical priority given that they increase the risk of perinatal morbidity and mortality as well as long-term diseases. A subset of small-for-gestational-age (SGA) infants are growth-restricted, and this condition is often attributed to placental insufficiency. Syndecan-1, a product of the degradation of the endothelial glycocalyx, has been proposed as a biomarker of endothelial damage in different pathologies. During pregnancy, a "specialized" form of the glycocalyx-the "syncytiotrophoblast glycocalyx"-covers the placental villi. The purpose of this study was to determine whether the concentration of maternal plasma syndecan-1 can be proposed as a biomarker for fetal growth restriction. STUDY DESIGN A cross-sectional study was designed to include women with normal pregnancy (n = 130) and pregnant women who delivered an SGA neonate (n = 50). Doppler velocimetry of the uterine and umbilical arteries was performed in women with an SGA fetus at the time of diagnosis. Venipuncture was performed within 48 h of Doppler velocimetry and plasma concentrations of syndecan-1 were determined by a specific and sensitive immunoassay. RESULTS (1) Plasma syndecan-1 concentration followed a nonlinear increase with gestational age in uncomplicated pregnancies (R2 = 0.27, p < .001); (2) women with a pregnancy complicated with an SGA fetus had a significantly lower mean plasma concentration of syndecan-1 than those with an appropriate-for-gestational-age fetus (p = .0001); (3) this difference can be attributed to fetal growth restriction, as the mean plasma syndecan-1 concentration was significantly lower only in the group of women with an SGA fetus who had abnormal umbilical and uterine artery Doppler velocimetry compared to controls (p = .00071; adjusted p = .0028). A trend toward lower syndecan-1 concentrations was also noted for SGA with abnormal uterine but normal umbilical artery Doppler velocimetry (p = .0505; adjusted p = .067); 4) among women with an SGA fetus, those with abnormal umbilical and uterine artery Doppler findings had a lower mean plasma syndecan-1 concentration than women with normal Doppler velocimetry (p = .02; adjusted p = .04); 5) an inverse relationship was found between the maternal plasma syndecan-1 concentration and the umbilical artery pulsatility index (r = -0.5; p = .003); and 6) a plasma syndecan-1 concentration ≤ 850 ng/mL had a positive likelihood ratio of 4.4 and a negative likelihood ratio of 0.24 for the identification of a mother with an SGA fetus who had abnormal umbilical artery Doppler velocimetry (area under the ROC curve 0.83; p < .001). CONCLUSION Low maternal plasma syndecan-1 may reflect placental diseases and this protein could be a biomarker for fetal growth restriction. However, as a sole biomarker for this condition, its accuracy is low.
Collapse
Affiliation(s)
- Alexander Juusela
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dahiana M Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Obstetrics and Gynecology, University del Valle, Cali, Colombia
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Obstetrics and Gynecology, AOUI Verona, University of Verona, Verona, Italy
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Adi L Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA.,Department of Computer Science, Wayne State University College of Engineering, Detroit, MI, USA
| | - Nandor Gabor Than
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary.,Maternity Private Clinic, Budapest, Hungary.,Department of Obstetrics and Gynecology, Semmelweis University, Budapest, Hungary
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.,Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA.,Detroit Medical Center, Detroit, MI, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI, USA.,Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|