1
|
Burdeau JA, Stephenson BJ, Aris IM, Mahalingaiah S, Chavarro JE, Preston EV, Hivert MF, Oken E, Calafat AM, Rifas-Shiman SL, Zota AR, James-Todd T. Early-pregnancy per- and polyfluoroalkyl substances and maternal post-pregnancy weight trajectory. Obesity (Silver Spring) 2025; 33:807-816. [PMID: 40074682 PMCID: PMC11975446 DOI: 10.1002/oby.24250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 03/14/2025]
Abstract
OBJECTIVE The objective of this study was to evaluate associations of early-pregnancy plasma per- and polyfluoroalkyl substances (PFAS) with maternal post-pregnancy weight trajectory parameters. METHODS We studied 1106 Project Viva participants with measures of early-pregnancy plasma concentrations of eight PFAS. We measured weight at in-person visits at 6 months and 3, 7, and 12 years after pregnancy and collected self-reported weight via annual questionnaires up to 17 years after pregnancy. Weight trajectory parameters were estimated via the Superimposition by Translation and Rotation model. We assessed individual and joint effects of PFAS with trajectory parameters using linear regression and Bayesian kernel machine regression (BKMR). RESULTS Perfluorooctane sulfonate (PFOS) concentrations were positively associated with weight trajectory magnitude in both linear regression (0.8 kg [95% CI: 0.1 to 1.4] per doubling of PFOS) and BKMR analyses (2.6 kg [95% CI: 1.4 to 3.8] per increase from 25th to 75th percentile of PFOS concentrations). Conversely, in BKMR analyses, perfluorononanoate was negatively associated with trajectory magnitude (-2.0 kg [95% CI: -2.9 to -1.1]). In stratified linear regression, older-aged participants had more pronounced positive associations of PFOS, perfluorooctanoate, and 2-(N-ethyl-perfluorooctane sulfonamido) acetate with weight trajectory velocity. No associations were observed with the overall PFAS mixture. CONCLUSIONS Select PFAS, assessed in pregnancy, may affect maternal weight trajectories spanning 17 years after pregnancy, especially for older-aged individuals.
Collapse
Affiliation(s)
- Jordan A. Burdeau
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Izzuddin M. Aris
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Shruthi Mahalingaiah
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - Jorge E. Chavarro
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Emma V. Preston
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Antonia M. Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Sheryl L. Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Ami R. Zota
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Tamarra James-Todd
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
2
|
Wei Y, Jiang X, Li H, Zhang Q, Hua L, Dong J, Xu J, Yang Y, Wang Q, Shen H, Zhang Y, Yan D, Peng Z, Kan H, Ma X, Cai J, He Y. Prenatal exposure to temperature variability, gestational duration and preterm birth: A nationwide birth cohort with 3 million singleton births in China. ENVIRONMENT INTERNATIONAL 2025; 198:109430. [PMID: 40209397 DOI: 10.1016/j.envint.2025.109430] [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: 08/24/2024] [Revised: 03/18/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Little is known about how temperature variability (TV) affects the risk of preterm birth (PTB). OBJECTIVE This cohort study aims to evaluate the associations of prenatal TV exposure with gestational age and PTB risk. METHODS This study included 3,012,744 live singleton births from 336 Chinese cities delivered between January 2013 and December 2015. TV exposure, indicating by standard deviation of temperature (SDT) and diurnal temperature range (DTR), were assessed by spatiotemporal models. Cox proportional hazard regression models were used to analyze the associations of PTB risk with TV exposure during the entire pregnancy and specific trimesters. Multivariate linear regressions were used to assess the associations between TV exposure and gestational age. RESULTS During the entire pregnancy, each 1 °C incremental in SDT and DTR was associated with a reduction in gestational age by 0.98 day (95 % confidence interval (CI), 0.89-1.06) and 0.36 day (95 % CI, 0.32-0.39). These increases in TV were also linked to a 20.1 % higher risk of PTB (hazard ratio [HR]: 1.201; 95 % CI: 1.182-1.233) for SDT and a 7.3 % higher risk of PTB (HR: 1.073; 95 % CI: 1.064-1.081) for DTR. The risk associated with TV was greater for very PTB than moderate and late PTB. Non-linear exposure-response curves indicated a monotonic increase in HRs for PTB with higher TV exposure, with the curves becoming less steep beyond an inflection point. These associations seemed to be more pronounced in women who lived in rural areas and in the climate zone of tropical monsoon, and gave birth in winter compared to their counterparts. CONCLUSIONS TV was associated with higher PTB risk and shortened gestational age in China.
Collapse
Affiliation(s)
- Yuhao Wei
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Xiaomin Jiang
- Department of Obstetrics, Anhui Province Hefei Maternity and Child Health Hospital, Hefei, Anhui 230001, China; Department of Obstetrics, Maternity and Child Healthcare Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230032, China
| | - Huimin Li
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qing Zhang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450053, China.
| | - Linlin Hua
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450053, China
| | - Jing Dong
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jihong Xu
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Ying Yang
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qiaomei Wang
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Haiping Shen
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yiping Zhang
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Donghai Yan
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Zuoqi Peng
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China
| | - Xu Ma
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jing Cai
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Fudan University, Shanghai 200032, China.
| | - Yuan He
- National Research Institute for Family Planning, Beijing 100081, China; National Human Genetic Resources Center, Beijing 101199, China; Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
3
|
Lopes NA, Ambeskovic M, King SE, Faraji J, Soltanpour N, Xu W, Fang X, Metz GAS, Olson DM. Transgenerational transmission of prenatal maternal stress across three generations of male progeny alters inflammatory stress markers in reproductive tissues. Psychoneuroendocrinology 2025; 177:107451. [PMID: 40179594 DOI: 10.1016/j.psyneuen.2025.107451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/18/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Prenatal maternal stress may lead to adverse pregnancy outcomes such as preterm birth and low birth weight. Our team has demonstrated in multiple rat models that prenatal maternal stress modifies the expression of inflammatory and stress regulators in the uterus and that this is transgenerationally passed over multiple generations through the female progeny. In this study, we investigated if male progeny exposed to ancestral prenatal maternal stress could also transmit changes to cause fetal programming of reproductive organs, leading to adverse pregnancy outcomes. We created a paternal transgenerational prenatal stress rat model. Dams (F0) were exposed to chronic variable stress during pregnancy, and their F1 male offspring stressed in utero were bred with control females for two generations. Gestational lengths and litter sizes were unchanged. Elevated gene expression of pro-inflammatory molecules in the uteri of F2 and F3 offspring was observed. Uterine expression of stress markers in the F2 and F3 females also increased even though plasma corticosterone levels were unchanged. Changes in the testicular expression of inflammatory and stress markers were also transmitted through the paternal lineage. These changes, however, tended to bear anti-inflammatory and adaptive functions, indicating compensatory mechanisms at play. These results demonstrate that fetal programming of uterine and testicular gene expression patterns can be transmitted through male progeny exposed to prenatal maternal stress.
Collapse
Affiliation(s)
- Nayara A Lopes
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Mirela Ambeskovic
- Canadian Centre for Behavioural Neuroscience and Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Stephanie E King
- Canadian Centre for Behavioural Neuroscience and Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Jamshid Faraji
- Canadian Centre for Behavioural Neuroscience and Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Nasrin Soltanpour
- Canadian Centre for Behavioural Neuroscience and Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Wendy Xu
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Xin Fang
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Gerlinde A S Metz
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Canadian Centre for Behavioural Neuroscience and Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada.
| | - David M Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Canadian Centre for Behavioural Neuroscience and Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; Department of Pediatrics, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
4
|
McIntyre TI, Valdez O, Kochhar NP, Davidson B, Samad B, Qiu L, Hu K, Combes AJ, Erlebacher A. KDM6B-dependent epigenetic programming of uterine fibroblasts in early pregnancy regulates parturition timing in mice. Cell 2025; 188:1265-1279.e18. [PMID: 39842437 PMCID: PMC11890963 DOI: 10.1016/j.cell.2024.12.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 09/03/2024] [Accepted: 12/13/2024] [Indexed: 01/24/2025]
Abstract
Current efforts investigating parturition timing mechanisms have focused on the proximal triggers of labor onset generated in late pregnancy. By studying the delayed parturition phenotype of mice with uterine fibroblast deficiencies in the histone H3K27me3 demethylase KDM6B, we provide evidence that parturition timing is regulated by events that take place in early pregnancy. Immediately after copulation, uterine fibroblasts engage in a locus-specific epigenetic program that abruptly adjusts H3K27me3 levels across their genome. In the absence of KDM6B, many of the adjusted loci over-accumulate H3K27me3. This over-accumulation leads to nearby genes being misexpressed in mid-to-late gestation, a delayed effect partly attributable to a second locus-specific but KDM6B-independent process initiated within uterine fibroblasts soon after implantation. This second process employs progressive H3K27me3 loss to temporally structure post-midgestational patterns of gene induction. Further dissection of the ways uterine programming controls parturition timing may have relevance to human pregnancy complications such as preterm labor.
Collapse
Affiliation(s)
- Tara I McIntyre
- Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Omar Valdez
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Nathan P Kochhar
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Brittany Davidson
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bushra Samad
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Longhui Qiu
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kenneth Hu
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexis J Combes
- UCSF CoLabs, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Division of Gastroenterology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Adrian Erlebacher
- Biomedical Sciences Program, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Bakar ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Center for Reproductive Science, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
5
|
Feyaerts D, Diop M, Galaz J, Einhaus JF, Arck PC, Diemert A, Winn VD, Parast M, Gyamfi-Bannerman C, Prins JR, Gomez-Lopez N, Stelzer IA. The single-cell immune profile throughout gestation and its potential value for identifying women at risk for spontaneous preterm birth. Eur J Obstet Gynecol Reprod Biol X 2025; 25:100371. [PMID: 40052005 PMCID: PMC11883378 DOI: 10.1016/j.eurox.2025.100371] [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: 07/30/2024] [Revised: 11/23/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Precisely timed immune adaptations, observed in the maternal circulation, underpin the notion of an immune clock of human pregnancy that supports its successful progression and completion at delivery. This immune clock is divided into three immunological phases, with the first phase starting at the time of conception and implantation, shifting into the second phase that supports homeostasis and tolerance throughout pregnancy, and culminating in the last phase of labor and parturition. Disruptions of this immune clock are reported in pregnancy complications such as spontaneous preterm birth. However, our understanding of the immune clock preceding spontaneous preterm birth remains scattered. In this review, we describe the chronology of maternal immune cell adaptations during healthy pregnancies and highlight its disruption in spontaneous preterm birth. With a focus on single-cell cytometric, proteomic and transcriptomic approaches, we review recent studies of term and spontaneous preterm pregnancies and discuss the need for future prospective studies aimed at tracking pregnancies longitudinally on a multi-omic scale. Such studies will be critical in determining whether spontaneous preterm pregnancies progress at an accelerated pace or follow a preterm-intrinsic pattern when compared to those delivered at term.
Collapse
Affiliation(s)
- Dorien Feyaerts
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Maïgane Diop
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jose Galaz
- Division of Obstetrics and Gynecology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jakob F. Einhaus
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Petra C. Arck
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anke Diemert
- Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mana Parast
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - Cynthia Gyamfi-Bannerman
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jelmer R. Prins
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nardhy Gomez-Lopez
- Departments of Obstetrics and Gynecology & Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Ina A. Stelzer
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
6
|
Dufford MT, Fleischer TC, Sommerville LJ, Badsha MB, Polpitiya AD, Logan J, Fox AC, Rust SR, Cox CB, Garite TJ, Boniface JJ, Kearney PE. Clock Proteins Have the Potential to Improve Term Delivery Date Prediction: A Proof-of-Concept Study. Life (Basel) 2025; 15:224. [PMID: 40003633 PMCID: PMC11856609 DOI: 10.3390/life15020224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/23/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Our ability to accurately predict the delivery date of term pregnancies is limited by shortcomings of modern-day clinical tools and due date estimation methods. The pregnancy clock is a series of coordinated and harmonized signals between mother, fetus, and placenta that regulate the length of gestation. Clock proteins are thought to be important mediators of these signals, yet few studies have investigated their potential utility as predictors of term delivery date. In this study, we performed a cross-sectional proteome analysis of 2648 serum samples collected between 18 and 28 weeks of gestation from mothers who delivered at term. The cohort included pregnancies both with and without complications. A total of 15 proteins of diverse functionalities were shown to have a direct association with time to birth (TTB), 11 of which have not been previously linked to gestational age. The protein A Distintegrin and Metalloproteinase 12 (ADA12) was one of the 15 proteins shown to have an association with TTB. Mothers who expressed the highest levels of ADA12 in the cohort (90th percentile) gave birth earlier than mothers who expressed the lowest levels of ADA12 (10th percentile) at a statistically significant rate (median gestational age at birth 390/7 weeks vs. 393/7 weeks, p < 0.001). Altogether, these findings suggest that ADA12, as well as potentially other clock proteins, have the potential to serve as clinical predictors of term delivery date in uncomplicated pregnancies and represent an important step towards characterizing the role(s) of clock proteins in mediating pregnancy length.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Paul E. Kearney
- Sera Prognostics, Inc., Salt Lake City, UT 84109, USA (T.C.F.); (A.C.F.); (T.J.G.)
| |
Collapse
|
7
|
Paquette A, Parenti M, Lapehn S, Konwar C, Kadam L, Firsick EJ, Barrett ES, MacIsaac J, MacDonald J, Bammler T, Carroll K, Enquobahrie D, Kobor M, LeWinn KZ, Nguyen R, Smith R, Spirzo A, Zhao Q, Myatt L, Bush NR, Muglia L, Sathyanarayana S. Associations between maternal plasma concentrations of corticotrophin releasing hormone and the placental transcriptome. Placenta 2025; 160:29-38. [PMID: 39755094 PMCID: PMC11980646 DOI: 10.1016/j.placenta.2024.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
INTRODUCTION The placenta produces corticotrophin releasing hormone (CRH), which rises exponentially in maternal plasma across pregnancy. CRH plays a functional role in fetal development, labor initiation, and the regulation of gestational length. We aimed to understand how maternal plasma CRH during pregnancy reflects placental physiology during parturition by characterizing placental transcriptomic signatures of maternal plasma CRH and comparing to transcriptomic signatures of gestational age at birth. METHODS Maternal plasma CRH concentrations were measured via radioimmunoassay at two timepoints and the placental transcriptome was quantified via RNA sequencing in 516 pregnant participants enrolled in the CANDLE cohort. Robust linear models were fitted to estimate associations between CRH and placental gene expression at birth. We conducted a functional validation in primary trophoblast cells before and after syncytialization. RESULTS Plasma CRH concentrations in the mid-pregnancy visit were associated with placental expression of 8 differentially expressed genes (DEGs), and concentrations in late pregnancy were associated with 283 DEGs. These genes were involved in several metabolic pathways. Seven genes were significantly associated with both plasma CRH and gestational length. Four genes were concordantly decreased and 7 genes were concordantly increased in primary trophoblasts treated with CRH. DISCUSSION Overall, this study reveals potential novel transcriptional mechanisms by which CRH may regulate metabolic pathways important for placental function and identifies genes associated with both CRH and gestational length.
Collapse
Affiliation(s)
- Alison Paquette
- Seattle Children's Research Institute, Seattle WA, USA; University of Washington, Seattle WA, USA.
| | | | | | - Chaini Konwar
- University of British Columbia, Vancouver BC, Canada
| | - Leena Kadam
- Oregon Health & Sciences University, Oregon WA, USA
| | | | - Emily S Barrett
- Rutgers University, Piscataway NJ, USA; University of Rochester, Rochester NY, USA
| | | | | | | | | | | | - Michael Kobor
- University of British Columbia, Vancouver BC, Canada
| | - Kaja Z LeWinn
- University of California San Francisco, San Francisco CA, USA
| | - Ruby Nguyen
- University of Minnesota, Minneapolis, MN, USA
| | - Roger Smith
- Hunter Medical Research Institute, University of Newcastle, Newcastle, USA
| | | | - Qi Zhao
- University of Tennessee Health Sciences Center, Memphis TN, USA
| | - Leslie Myatt
- Oregon Health & Sciences University, Oregon WA, USA
| | - Nicole R Bush
- University of California San Francisco, San Francisco CA, USA
| | - Louis Muglia
- The Burroughs Wellcome Fund, Research Triangle Park NC, USA
| | - Sheela Sathyanarayana
- Seattle Children's Research Institute, Seattle WA, USA; University of Washington, Seattle WA, USA
| |
Collapse
|
8
|
Mayne G, DeWitt PE, Wen J, Schniedewind B, Dabelea D, Christians U, Hurt KJ. Adiponectin and Glucocorticoids Modulate Risk for Preterm Birth: The Healthy Start Study. J Clin Endocrinol Metab 2025; 110:523-533. [PMID: 38980936 DOI: 10.1210/clinem/dgae464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/18/2024] [Accepted: 07/05/2024] [Indexed: 07/11/2024]
Abstract
CONTEXT Adiponectin is a potent uterine tocolytic that decreases with gestational age, suggesting it could be a maternal metabolic quiescence factor. Maternal stress can influence preterm birth risk, and adiponectin levels may be stress responsive. OBJECTIVE We characterized associations between adiponectin and glucocorticoids with preterm birth and modeled their predictive utility. We hypothesized maternal plasma adiponectin and cortisol are inversely related and lower adiponectin and higher cortisol associate with preterm birth. METHODS We performed a nested case-control study using biobanked fasting maternal plasma. We included low-risk singleton pregnancies, and matched 1:3 (16 preterm, 46 term). We quantified high molecular weight (HMW), low molecular weight (LMW), and total adiponectin using an enzyme-linked immunosorbent assay. We validated a high-performance liquid chromatography-tandem mass spectrometry serum assay for use in plasma, to simultaneously measure cortisol, cortisone, and 5 related steroid hormones. We used linear/logistic regression to compare group means and machine learning for predictive modeling. RESULTS The preterm group had lower mean LMW adiponectin (3.07 μg/mL vs 3.81 μg/mL at 15 weeks (w) 0 days (d), P = .045) and higher mean cortisone (34.4 ng/mL vs 29.0 ng/mL at 15w0d, P = .031). The preterm group had lower cortisol to cortisone and lower LMW adiponectin to cortisol ratios. We found HMW adiponectin, cortisol to cortisone ratio, cortisone, maternal height, age, and prepregnancy body mass index most strongly predicted preterm birth (area under the receiver operator curve = 0.8167). In secondary analyses, we assessed biomarker associations with maternal self-reported psychosocial stress. Lower perceived stress was associated with a steeper change in cortisone in the term group. CONCLUSION Overall, metabolic and stress biomarkers are associated with preterm birth in this healthy cohort. We identify a possible mechanistic link between maternal stress and metabolism for pregnancy maintenance.
Collapse
Affiliation(s)
- Gabriella Mayne
- Department of Health & Behavioral Sciences, University of Colorado, Denver, CO 80204, USA
| | - Peter E DeWitt
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jennifer Wen
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Björn Schniedewind
- iC42 Clinical Research & Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dana Dabelea
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Uwe Christians
- iC42 Clinical Research & Development, Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K Joseph Hurt
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
9
|
Georges HM, Norwitz ER, Abrahams VM. Predictors of Inflammation-Mediated Preterm Birth. Physiology (Bethesda) 2025; 40:0. [PMID: 39106300 DOI: 10.1152/physiol.00022.2024] [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: 04/30/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 08/09/2024] Open
Abstract
Preterm birth remains a worldwide health concern because of ongoing challenges in prediction and prevention. Current predictors are limited by poor performance, need for invasive sampling, and an inability to identify patients in a timely fashion to allow for effective intervention. The multiple etiologies of preterm birth often have an inflammatory component. Thus, a deeper understanding of the inflammatory mechanisms involved in preterm birth may provide opportunities to identify new predictors of preterm birth. This review discusses the multiple etiologies of preterm birth, their links to inflammation, current predictors available, and new directions for the field.
Collapse
Affiliation(s)
- Hanah M Georges
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
| | - Errol R Norwitz
- Department of Obstetrics and Gynecology, Tufts University School of Medicine, Boston, Massachusetts, United States
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
10
|
Wang B, Seif K, Lei J, Mangione M, Turan S, Reece EA, Burd I, Yang P. Novel Placental Biomarker Shows Predictive Potential for Spontaneous Preterm Labor. Am J Perinatol 2024. [PMID: 39613308 DOI: 10.1055/a-2491-4391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
OBJECTIVE Human parturition involves many events among mother, fetus, and placenta, and the initiation of these events is the consequence of activation of a series of endocrine and immune responses. Multiple underlying pathways account for the cascade of events that culminate in spontaneous preterm labor. In this study, we aimed to characterize these signaling pathways of placental origin at molecular levels. STUDY DESIGN We used single-cell RNA-sequencing (sc-RNA-seq) analysis to probe transcriptional heterogeneity in human placenta delivered at preterm or term and then used RNA in situ hybridization (RNA-ISH) assay on formalin-fixed paraffin-embedded (FFPE) placental tissues to validate these results. RESULTS By using sc-RNA-seq on villous cytotrophoblast (CTB) isolated from a preterm placenta, we found that signaling pathways implicated in the initiation of term or preterm labor including ferroptosis, kisspeptin (KISS1), and senescence were constitutively activated in distinct cellular clusters of these trophoblastic stem cells. RNA-ISH-based spatial gene expression profiling in FFPE tissues revealed that pregnancy-specific beta-1-glycoprotein 4 (PSG4), a potent molecular driver for cellular aging, was significantly increased in preterm placentas (N = 30) compared to their full-term counterparts (N = 9). Furthermore, PSG4 mRNA signals were predominantly detected in the villous syncytiotrophoblast and the discontinuous monolayer of CTB as well as the intervillous space where maternal blood circulates. CONCLUSION Our study provides strong support for PSG4 overexpression serving as a biomarker for pregnant women at risk for preterm delivery, which can allow for the development of timely and clinical preventive strategies. KEY POINTS · A sc-RNA-seq analysis on a preterm placenta identified multiple signaling pathways constitutively activated in distinct clusters of CTB.. · RNA-ISH assay uncovered that the mRNA levels of PSG4, a molecular driver for cellular senescence significantly increased in preterm placentas compared to the term counterparts.. · PSG4 mRNA signals were largely observed in the villous trophoblast as well as the intervillous space..
Collapse
Affiliation(s)
- Bingbing Wang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Karl Seif
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jun Lei
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Mary Mangione
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sifa Turan
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - E Albert Reece
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Irina Burd
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Pande A, Kinkade CW, Prout N, Chowdhury SF, Rivera-Núñez Z, Barrett ES. Prenatal exposure to synthetic chemicals in relation to HPA axis activity: A systematic review of the epidemiological literature. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 956:177300. [PMID: 39488279 DOI: 10.1016/j.scitotenv.2024.177300] [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: 08/28/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Pregnant people are widely exposed to numerous synthetic chemicals with known endocrine-disrupting properties (e.g., phthalates, phenols, per- and poly-fluoroalkyl substances (PFAS)). To date, most epidemiological research on how endocrine-disrupting chemicals (EDCs) disrupt hormone pathways has focused on estrogens, androgens, and thyroid hormones. Far less research has examined the impact of EDCs on the hypothalamic-pituitary-adrenal (HPA) axis, despite its central role in the physiologic stress response and metabolic function. OBJECTIVE To systematically review the epidemiological literature on prenatal synthetic EDC exposures in relation to HPA axis hormones (e.g., corticotropin-releasing hormone, adrenocorticotropic hormone, cortisol, cortisone) in pregnant people and their offspring. METHODS A literature search of PubMed, Scopus, and Embase was conducted. Primary research studies were selected for inclusion by two independent reviewers and risk of bias was assessed using the Office of Health Assessment and Translation guidelines established by the National Toxicology Program with customization for the specific research topic. Data were extracted from each study and included in a qualitative synthesis. RESULTS 22 published studies met the inclusion criteria. Phthalates were the most prevalent EDC studied, followed by PFAS, phenols, and parabens, with fewer studies considering other synthetic chemicals. Offspring glucocorticoids were the most commonly considered outcome, followed by maternal glucocorticoids and placental corticotropin-releasing hormone. There was considerable heterogeneity in methods across studies, particularly in HPA axis outcome measures and matrices, making cross-study comparisons challenging. Numerous studies suggested disruption of HPA axis hormones and sex differences in association, but results varied considerably across studies and EDC classes. CONCLUSIONS The limited literature to date suggests the HPA axis may be vulnerable to disruption by synthetic EDCs. Carefully designed studies that prioritize biospecimen collection specific to HPA axis hormones are needed along with greater standardization of biospecimen collection and analysis protocols to facilitate cross-study comparisons and interpretation.
Collapse
Affiliation(s)
- Anushka Pande
- Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | - Carolyn W Kinkade
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA
| | - Nashae Prout
- Wynne Center for Family Research, University of Rochester, Rochester, NY 14642, USA; Department of Environmental Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14620, USA
| | - Sadia F Chowdhury
- Wynne Center for Family Research, University of Rochester, Rochester, NY 14642, USA; Translational Biomedical Sciences Program, University of Rochester, Rochester, NY 14642, USA
| | - Zorimar Rivera-Núñez
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA; Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ 08854, USA
| | - Emily S Barrett
- Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ 08854, USA; Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ 08854, USA.
| |
Collapse
|
12
|
Catalano RA, Bruckner TA, Gemmill A, Margerison CE. The pandemic preterm paradox: a test of competing explanations. Am J Epidemiol 2024; 193:1823-1831. [PMID: 38885959 PMCID: PMC11637531 DOI: 10.1093/aje/kwae128] [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: 05/19/2023] [Revised: 04/20/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
Epidemiologists have long argued that side effects of the stress response include preterm birth. Research reports that fear of lethal infection stressed pregnant persons at the outset of the coronavirus disease (COVID-19) pandemic and that "shutdowns" and "social distancing" impeded access to social support and prenatal care. The decline in preterm births in high-income countries, including the United States, during the early months of the pandemic therefore poses a paradox for science. Explanations of this "pandemic preterm paradox" remain untested. We applied time-series modeling to data describing 80 monthly conception cohorts begun in the United States from July 2013 through February 2020 to determine which of 3 explanations most parsimoniously explained the paradox. We infer that "prior loss," or the argument that an increase in spontaneous abortions and stillbirths depleted the population of fetuses at risk of preterm birth, best explains data currently available. We describe the implications of these results for public health practice.
Collapse
Affiliation(s)
- Ralph A Catalano
- School of Public Health, University of California, Berkeley, CA 94704, United States
| | - Tim A Bruckner
- Joe C. Wen School of Population & Public Health and Center for Population, Inequality and Policy, University of California, Irvine, CA 92697, United States
| | - Alison Gemmill
- Department of Population, Family and Reproductive Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Claire E Margerison
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI 48824, United States
| |
Collapse
|
13
|
Chen Y, Künzel RG, Sanchez SE, Rondon MB, Pinto NI, Sanchez E, Kirschbaum C, Valeri L, Koenen KC, Gelaye B. The association between pre-pregnancy and first-trimester hair cortisol and preterm birth: a causal inference model. Eur J Epidemiol 2024; 39:1391-1400. [PMID: 39661097 PMCID: PMC11680651 DOI: 10.1007/s10654-024-01174-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/27/2024] [Indexed: 12/12/2024]
Abstract
Adverse life events and chronic psychological distress before and during pregnancy have frequently been associated with preterm birth but the biological underpinnings remain unclear. We investigated the association between corticosteroid levels in pre-pregnancy and first-trimester hair and the risk of preterm birth. We followed N = 1,807 pregnant women from a prospective pre-birth cohort study in Lima, Perú. Hair samples were taken at the end of the first pregnancy trimester. The two most proximal 3 cm segments to the scalp (representing pre-pregnancy and first-trimester) were analyzed to obtain hair cortisol and cortisone concentrations (HCC and HCNC). Preterm birth was defined as birth < 37 completed gestational weeks. We constructed four generalized propensity scores for pre-pregnancy and first-trimester HCC and HCNC to create corresponding inverse probability weights before fitting marginal structural models for estimating the effect of HCC and HCNC on preterm birth risk. Pre-pregnancy Log HCC was not independently associated with preterm birth risk (RR = 0.97; 95%CI: 0.79, 1.19), nor was pre-pregnancy Log HCNC (RR = 0.84; 95%CI: 0.58, 1.20). In the first trimester, a one SD increase in Log HCC was associated with a 37% increased risk of preterm birth (95%CI: 1.11, 1.69), whereas Log HCNC was not significantly associated with preterm birth risk (RR = 1.20; 95%CI: 0.87, 1.65). Our findings show that chronic corticosteroid levels in early pregnancy are causally linked to preterm birth risk in pregnant Peruvian women. This finding contributes to understanding the biological underpinnings of preterm birth better to enhance its prevention.
Collapse
Affiliation(s)
- Yinxian Chen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Epidemiology, Rollins School of Public Health, Emory University, GA, 30322, Atlanta, USA
| | - Richard G Künzel
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
- Katholische Universität Eichstätt-Ingolstadt, 85072, Eichstätt, Germany.
| | - Sixto E Sanchez
- Facultad de Medicina Humana, Universidad de San Martin de Porres, Instituto de Investigación, Lima, 15024, Perú
- Asociación Civil Proyectos en Salud, Lima, 15024, Perú
| | | | - Nelida I Pinto
- Facultad de Medicina Humana, Universidad de San Martin de Porres, Instituto de Investigación, Lima, 15024, Perú
| | - Elena Sanchez
- Asociación Civil Proyectos en Salud, Lima, 15024, Perú
| | | | - Linda Valeri
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY, 10032, USA
| | - Karestan C Koenen
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Bizu Gelaye
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Psychiatry, The Chester M. Pierce M.D. Division of Global Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
14
|
Nu UT, Pervin J, Rahman M, Kamal KTB, Aktar S, Huda FA, Ganguly S, El Arifeen S, Persson LÅ, Rahman A. A cohort study of the occurrence of post-term births and its association with perinatal mortality in a rural area in Bangladesh. J Glob Health 2024; 14:04238. [PMID: 39541503 PMCID: PMC11565093 DOI: 10.7189/jogh.14.04238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Background We aimed to evaluate the trend of post-term births over time and their association with perinatal mortality based on prospective pregnancy cohorts in a rural area in Bangladesh. Methods This cohort study included 72 373 singleton births with gestational ages ≥28 weeks recorded by a health and demographic surveillance system from 1990 to 2019 in Matlab, Bangladesh. We expressed the gestational age as X (weeks) + Y (days)/7 weeks, where X indicated complete weeks, and Y presented the number of completed days out of seven days or a week. Using Poisson regression with robust variances, we estimated the population-based proportion of post-term births and assessed the association between gestational age categories and perinatal mortality. We presented results by adjusted relative risk (aRR) with 95% confidence intervals (CIs). Results Post-term births declined from 5.8% in 1990 to 2.8% in 2019. Perinatal mortality declined from 58 to 27 per 1000 births from 1990 to 2019. Compared to full-term births (39 + 0/7 to 40 + 6/7 weeks), the aRRs of perinatal mortality were 1.39 for late-term (41 + 0/7 to 41 + 6/7 weeks) and 1.93 for post-term (≥42 + 0/7 weeks) births. The population-attributable fraction of perinatal mortality was 15% for births at ≥41 + 0/7 weeks out of the total perinatal deaths occurring at ≥39 gestation weeks. Conclusions In this rural setting in Bangladesh, we observed a decline in post-term birth proportions from 1990 to 2019. We found increased perinatal mortality when pregnancy continued beyond 40 + 6/7 weeks of gestation. This implies that planning the management of pregnant women approaching the post-term period may be needed to further improve perinatal health outcomes.
Collapse
Affiliation(s)
- U Tin Nu
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Jesmin Pervin
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Monjur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Kazi Tamara B Kamal
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shaki Aktar
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Fauzia A Huda
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Shikha Ganguly
- Department of Gynaecology and Obstetrics, Dhaka Medical College and Hospital, Dhaka, Bangladesh
| | - Shams El Arifeen
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Lars Åke Persson
- Department of Disease Control, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Anisur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| |
Collapse
|
15
|
Deer LK, Demers CH, Hankin BL, Doom JR, Shields GS, Hoffman MC, Davis EP. Neonatal Hair Cortisol and Birth Outcomes: An Empirical Study and Meta-Analysis. Psychosom Med 2024; 86:720-729. [PMID: 39132972 DOI: 10.1097/psy.0000000000001339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
OBJECTIVE Prenatal stress physiology is often posited as a predictor of birth outcomes, including gestational age at birth and birthweight. However, research has predominantly relied on indicators in the maternal system, with few studies examining hormones of the fetal system. The current study focuses on fetal cortisol in the third trimester, as measured in neonatal hair, as a biological factor that might be associated with birth outcomes (gestational age at birth and birthweight). We report findings from two studies: a longitudinal cohort (Study 1), and a meta-analysis of the existing literature (Study 2). METHODSSTUDY Hair was collected for cortisol analysis from 168 neonates (55.95% female) shortly after birth. Gestational age at birth and birthweight were abstracted from medical records. METHODSSTUDY An exhaustive search of four databases was conducted, yielding 155 total studies for screening. Papers reporting neonatal hair cortisol (collection <2 weeks postpartum) and birth outcomes among human neonates were retained for analysis, including Study 1 results ( k = 9). RESULTSSTUDY Higher neonatal hair cortisol was related to longer gestation ( r = 0.28, p < .001) and higher birthweight, r = 0.16, p = .040. Sex did not moderate either association. RESULTSSTUDY Across the nine studies, higher neonatal hair cortisol predicted both longer gestation ( r = 0.35, p < .001, 95% confidence interval = 0.24-0.45) and higher birthweight ( r = 0.18, p = .001, 95% confidence interval = 0.07-0.28). Neonatal sex did not moderate these associations. CONCLUSIONS Fetal cortisol exposure in the third trimester plays a role in normative maturation of the fetus, and findings reveal that higher cortisol is associated with positive birth outcomes.
Collapse
Affiliation(s)
- LillyBelle K Deer
- From the Department of Psychology (Deer, Doom, Davis), University of Denver, Denver; Department of Psychiatry (Demers, Hoffman), University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Psychology (Hankin), University of Illinois Urbana-Champaign, Champaign, Illinois; Department of Psychological Science (Shields), University of Arkansas, Fayetteville, Arkansas; Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology (Hoffman), University of Colorado Anschutz Medical Campus, Aurora, Colorado; and Department of Pediatrics (Davis), University of California, Irvine, Irvine, California
| | | | | | | | | | | | | |
Collapse
|
16
|
Juvinao-Quintero DL, Künzel RG, Larrabure-Torrealva G, Duncan L, Kirschbaum C, Sanchez SE, Gelaye B. Characterization of hair cortisol concentration pre-conception and during pregnancy. Psychoneuroendocrinology 2024; 167:107089. [PMID: 38850884 PMCID: PMC11328972 DOI: 10.1016/j.psyneuen.2024.107089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/19/2024] [Accepted: 05/30/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND The hypothalamic pituitary adrenal (HPA) axis is a system involved in stress and pregnancy regulation, and hair cortisol concentration (HCC) is a promising biomarker of its activity. Assessing factors that influence HCC in the prenatal period is critical to understand whether and how HPA axis (dys-)regulation influences maternal health and child development, particularly in high-risk populations from low- and middle-income countries (LMICs). AIMS This study aimed at characterizing preconception and pregnancy HCC with respect to multiple sociodemographic, pregnancy-related, and hair-related factors. METHODS In a sample of N = 2581 pregnant women in Perú, participants from two cohort studies provided a 6 cm scalp hair sample at three prenatal timepoints. Each hair sample was cut into two segments of 3 cm that represent cortisol secretion at four times: preconception, first-, second- and third trimester of pregnancy. Hair cortisol was extracted using liquid chromatography tandem mass spectrometry (LC-MS/MS). Spearman correlations, paired t-tests, and ANOVA were used to assess differences in log-transformed values of HCC (logHCC) across maternal sociodemographic, pregnancy-related, and hair-related factors. Multivariable linear regressions were used to examine independent associations of HCCs with selected correlates. RESULTS Mean logHCC values showed an increase across the four prenatal periods. Preconception BMI was consistently associated with HCC in all three trimesters, while difficulty accessing basic foods, education, hair dyeing, and infant sex showed time-specific associations with HCCs. In sensitivity analyses, we detected no substantial segment effects in the associations of HCCs with maternal characteristics. CONCLUSION This study is the largest to characterize HCC in pregnant women from a LMIC. Our findings provide a foundation for the use of HCC as a biomarker of prenatal HPA axis activity for future studies. This foundation may contribute to finding valid biomarkers of stress-response systems to promote maternal and child health.
Collapse
Affiliation(s)
- Diana L Juvinao-Quintero
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, 677 Huntington Avenue, Kresge 500, Boston, MA 02115, USA
| | - Richard G Künzel
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, 677 Huntington Avenue, Kresge 500, Boston, MA 02115, USA; Katholische Universität Eichstätt-Ingolstadt, Ostenstraße 26, Eichstätt 85072, Germany.
| | - Gloria Larrabure-Torrealva
- Instituto Nacional Materno Perinatal, Lima 15024, Peru; Universidad Nacional Mayor de San Marcos, Instituto de Investigaciones Clínicas, Lima 15081, Peru
| | - Laramie Duncan
- Stanford University, Department of Psychiatry and Behavioral Sciences, 401 Quarry Road, Stanford, CA 94305, USA
| | - Clemens Kirschbaum
- Technische Universität Dresden, Zellescher Weg 19, Dresden 01069, Germany
| | - Sixto E Sanchez
- Universidad de San Martin de Porres, Facultad de Medicina Humana, Instituto de Investigacion, Lima 15024, Peru; Asociación Civil PROESA, Lima 15024, Peru
| | - Bizu Gelaye
- Harvard T.H. Chan School of Public Health, Department of Epidemiology, 677 Huntington Avenue, Kresge 500, Boston, MA 02115, USA; Massachusetts General Hospital and Harvard Medical School, The Chester M. Pierce, M.D. Division of Global Psychiatry, 151 Merrimac Street, Boston, MA 02114, USA
| |
Collapse
|
17
|
Hamburg-Shields E, Mesiano S. The hormonal control of parturition. Physiol Rev 2024; 104:1121-1145. [PMID: 38329421 PMCID: PMC11380996 DOI: 10.1152/physrev.00019.2023] [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: 04/20/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/09/2024] Open
Abstract
Parturition is a complex physiological process that must occur in a reliable manner and at an appropriate gestation stage to ensure a healthy newborn and mother. To this end, hormones that affect the function of the gravid uterus, especially progesterone (P4), 17β-estradiol (E2), oxytocin (OT), and prostaglandins (PGs), play pivotal roles. P4 via the nuclear P4 receptor (PR) promotes uterine quiescence and for most of pregnancy exerts a dominant block to labor. Loss of the P4 block to parturition in association with a gain in prolabor actions of E2 are key transitions in the hormonal cascade leading to parturition. P4 withdrawal can occur through various mechanisms depending on species and physiological context. Parturition in most species involves inflammation within the uterine tissues and especially at the maternal-fetal interface. Local PGs and other inflammatory mediators may initiate parturition by inducing P4 withdrawal. Withdrawal of the P4 block is coordinated with increased E2 actions to enhance uterotonic signals mediated by OT and PGs to promote uterine contractions, cervix softening, and membrane rupture, i.e., labor. This review examines recent advances in research to understand the hormonal control of parturition, with focus on the roles of P4, E2, PGs, OT, inflammatory cytokines, and placental peptide hormones together with evolutionary biology of and implications for clinical management of human parturition.
Collapse
Affiliation(s)
- Emily Hamburg-Shields
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, Ohio, United States
- Department of Obstetrics and Gynecology, University Hospitals of Cleveland, Cleveland, Ohio, United States
| | - Sam Mesiano
- Department of Reproductive Biology, Case Western Reserve University, Cleveland, Ohio, United States
- Department of Obstetrics and Gynecology, University Hospitals of Cleveland, Cleveland, Ohio, United States
| |
Collapse
|
18
|
Künzel RG, Elgazzar M, Bain PA, Kirschbaum C, Papatheodorou S, Gelaye B. The association between maternal prenatal hair cortisol concentration and preterm birth: A systematic review and meta-analysis. Psychoneuroendocrinology 2024; 165:107041. [PMID: 38581747 DOI: 10.1016/j.psyneuen.2024.107041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/30/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND The risk of preterm birth (PTB) increases when experiencing stress during pregnancy. Chronic stress has been associated with a dysregulation of the hypothalamic-pituitary-adrenal axis, for which hair cortisol concentration (HCC) is a promising biomarker. However, previous studies on the association between HCC and PTB yielded inconsistent results. This systematic review and meta-analysis synthesized previous studies on the association between maternal HCC before and during pregnancy and spontaneous PTB. METHODS Data was extracted from N = 11 studies with k = 19 effect sizes retrieved from PubMed, Embase, Web of Science, CINAHL and citation searching by hand in June 2023 and updated in October 2023. Standardized mean differences were calculated, and a random-effects three-level meta-analysis was conducted. Effect heterogeneity was assessed using Q and I2. RESULTS HCC during pregnancy was higher among PTB than term groups, but effects were not statistically significant (z = 0.11, 95% CI: - 0.28, 0.51, p = .54) and total heterogeneity was high (Q16 = 60.01, p < .001, I2Total = 92.30%). After leaving out two possible outlier studies in sensitivity analyses, HCC was lower among preterm compared to term delivering groups, although not statistically significant (z = - 0.06, 95% CI: - 0.20, 0.08, p = .39) but with a substantially reduced total heterogeneity (Q12 = 16.45, p = .17, I2Total = 42.15%). No moderators affected the estimates significantly, but an effect of trimester and gestational age at delivery is likely. CONCLUSION There is currently no evidence of prenatal HCC differences between PTB and term groups as effects were small, imprecise, and not significant. Low statistical power and methodological weaknesses of the small-scale studies challenge possible biological inferences from the small effects, but further research on HCC during pregnancy is highly encouraged.
Collapse
Affiliation(s)
- Richard G Künzel
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA; Katholische Universität Eichstätt-Ingolstadt, Ostenstr. 28a, Eichstätt 85072, Germany.
| | | | - Paul A Bain
- Countway Library, Harvard Medical School, 10 Shattuck St, Boston, MA 02115, USA
| | - Clemens Kirschbaum
- Technische Universität Dresden, Zellescher Weg 19, Dresden 01062, Germany
| | - Stefania Papatheodorou
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA
| | - Bizu Gelaye
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Avenue, Boston, MA 02115, USA; The Chester M. Pierce M.D. Division of Global Psychiatry, Department of Psychiatry, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114, USA
| |
Collapse
|
19
|
Wada Y, Watanabe J, Yamamoto N, Kubota T, Kamijo K, Hirano D, Takahashi H, Fujiwara H. Association between earthquakes and perinatal outcomes: A systematic review and meta-analysis. Int J Gynaecol Obstet 2024; 166:228-237. [PMID: 38234161 DOI: 10.1002/ijgo.15369] [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: 06/23/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024]
Abstract
BACKGROUND Evidence for the association between earthquakes and adverse perinatal outcomes is limited. OBJECTIVES To evaluate the association between earthquakes and perinatal outcomes including preterm birth and low birth weight. SEARCH STRATEGY We searched studies using MEDLINE, Cochrane Central Register of Controlled Trials, EMBASE, World Health Organization International Clinical Trials Platform Search Portal, and ClinicalTrials.gov on February 9, 2023. SELECTION CRITERIA We included before-and-after studies that evaluated the associations between earthquakes and perinatal outcomes in women living in affected areas. DATA COLLECTION AND ANALYSIS Two independent reviewers extracted data. We calculated the pooled odds ratio (OR) with the random-effects model. We analyzed outcomes in subgroups of Asians and others. We evaluated the certainty of evidence with the Grading of Recommendations Assessment, Development and Evaluation system. MAIN RESULTS We included 2 607 405 women in 13 studies. Earthquakes may not increase preterm birth (nine studies, 1 761 760 participants: OR 1.10, 95% confidence interval [CI] 0.98-1.24, low certainty of evidence) or low birth weight (seven studies, 1 753 891 participants: OR 1.10, 95% CI 0.94-1.28, low certainty of evidence). Subgroup analyses showed that earthquakes may be associated with an increase of preterm birth among populations in Asia (OR 1.44, 95% CI 1.07-1.95), but this was not evident in others (OR 0.93, 95% CI 0.83-1.05). CONCLUSIONS Perinatal outcomes might not change after earthquakes. Further research on the association between earthquakes and perinatal outcomes, combined with an assessment of the characteristics of the region, is needed.
Collapse
Affiliation(s)
- Yoshimitsu Wada
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
- Scientific Research WorkS Peer Support Group (SRWS-PSG), Osaka, Japan
| | - Jun Watanabe
- Scientific Research WorkS Peer Support Group (SRWS-PSG), Osaka, Japan
- Department of Surgery, Division of Gastroenterological, General and Transplant Surgery, Jichi Medical University, Tochigi, Japan
- Center for Community Medicine, Jichi Medical University, Tochigi, Japan
| | - Norio Yamamoto
- Scientific Research WorkS Peer Support Group (SRWS-PSG), Osaka, Japan
- Department of Epidemiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takafumi Kubota
- Scientific Research WorkS Peer Support Group (SRWS-PSG), Osaka, Japan
- Department of Neurology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kyosuke Kamijo
- Scientific Research WorkS Peer Support Group (SRWS-PSG), Osaka, Japan
- Department of Gynecology, Nagano Municipal Hospital, Nagano, Japan
| | - Daishi Hirano
- Scientific Research WorkS Peer Support Group (SRWS-PSG), Osaka, Japan
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Hironori Takahashi
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| | - Hiroyuki Fujiwara
- Department of Obstetrics and Gynecology, Jichi Medical University, Tochigi, Japan
| |
Collapse
|
20
|
Almeida IF, Rinne GR, Coussons-Read M, Dunkel Schetter C. Placental corticotrophin-releasing hormone trajectories in pregnancy: Associations with postpartum depressive symptoms. Psychoneuroendocrinology 2024; 164:107030. [PMID: 38537413 PMCID: PMC11373738 DOI: 10.1016/j.psyneuen.2024.107030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/07/2024] [Accepted: 03/17/2024] [Indexed: 04/21/2024]
Abstract
OBJECTIVE Depressive symptoms following birth are common and can have adverse effects for mothers, children, and families. Changes in hypothalamic-pituitary-adrenal (HPA) axis regulation during pregnancy may be implicated in the development of postpartum depressive symptoms, particularly changes in placental corticotropinreleasing hormone (pCRH). However, few studies have tested how dynamic pCRH changes over pregnancy relate to postpartum depressive symptoms. This preregistered investigation tests associations of both pCRH levels and changes from early to late pregnancy with postpartum depressive symptoms. METHODS The sample consists of 173 women studied in early, mid, and late pregnancy who later reported on depressive symptoms with the Edinburgh Postpartum Depression Scale during interviews at 1, 6 and 12 months postpartum. Blood samples were collected at each prenatal timepoint and assayed for pCRH using radioimmunoassay. Latent growth curve analysis was employed to identify distinct trajectories of pCRH during pregnancy. RESULTS We identified three prenatal pCRH trajectories labeled as typical, flat, and accelerated. Each trajectory showed exponential increases in pCRH levels over the course of gestation but differed in overall levels and rates of change. pCRH levels were not associated with postpartum depressive symptoms. However, women with accelerated pCRH trajectories reported marginally higher depressive symptoms one month postpartum. Primary analysis models adjusted for marital status, income, prepregnancy BMI, parity, prenatal depressive symptoms, and gestational age. CONCLUSIONS These findings add to our understanding of dynamic changes to maternal HPA axis regulation during pregnancy and contribute to growing evidence on how pCRH changes relate to the development of postpartum depressive symptoms.
Collapse
|
21
|
Chai Y, Wang H, Tang D, Wu Y, Sun Z, Zeng Y, Zhang B, Niu B, Dong X. Changes of serum cortisol during pregnancy and labor initiation: an onsite cross-sectional study. Front Endocrinol (Lausanne) 2024; 15:1379693. [PMID: 38808114 PMCID: PMC11130462 DOI: 10.3389/fendo.2024.1379693] [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: 01/31/2024] [Accepted: 04/15/2024] [Indexed: 05/30/2024] Open
Abstract
Background Increased maternal cortisol secretion has been observed during pregnancy and labor. However, due to the limitations in diagnostic methods, the dynamic change of cortisol during the short period between threatened labor and labor is unknown. In this study, we aim to evaluate the changes in serum cortisol during late pregnancy and full-term labor initiation, verifying if cortisol could serve as a biomarker for the diagnosis of labor initiation from threatened labor. Methods This cross-sectional onsite study involved 564 participants of 6 different gestational stages (C: Control; T1: Trimester 1; T3: Trimester 3; E: expectant; TL: threatened labor; L: labor), all patients in the E, TL, and L groups were at full term. The serum cortisol concentration was quantified with a point-of-care test (POCT), and the gestation, age, parity, and BMI of participants were documented. Morning serum cortisol was collected between 8:00 and 10:00 a.m., except for the TL and L group women who were tested upon arrival or during latent labor. With cortisol levels or all five variables, L was distinguished from TL using machine learning algorithms. Results Significant elevation of cortisol concentration was observed between T1 and T3, or TL and L group (P< 0.001). Women belonging to the E and TL group showed similar gestation week and cortisol levels. Diagnosis of labor initiation using cortisol levels (cutoff = 21.46 μg/dL) yielded sensitivity, specificity, and AUC of 86.50%, 88.60%, and 0.934. With additional variables, a higher specificity (89.29%) was achieved. The diagnostic accuracy of all methods ranged from 85.93% to 87.90%. Conclusion Serum cortisol could serve as a potential biomarker for diagnosis of L form TL. The rapid onsite detection of serum cortisol with POCT could facilitate medical decision-making for admission and special treatments, either as an additional parameter or when other technical platforms are not available.
Collapse
Affiliation(s)
- Yujuan Chai
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - Hairong Wang
- Greater Bay Area International Institute for Innovation, Shenzhen University, Shenzhen, Guangdong, China
| | - Daiyu Tang
- Department of Obstetrics and Gynecology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| | - Yi Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhonghao Sun
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - Yuping Zeng
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - Binmao Zhang
- Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong, China
| | - Ben Niu
- Department of Management, Shenzhen University, Shenzhen, Guangdong, China
| | - Xiaojing Dong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
22
|
Barrett ES, Sullivan A, Workman T, Zhang Y, Loftus CT, Szpiro AA, Paquette A, MacDonald JW, Coccia M, Smith R, Bowman M, Smith A, Derefinko K, Nguyen RHN, Zhao Q, Sathyanarayana S, Karr C, LeWinn KZ, Bush NR. Sex-specific associations between placental corticotropin releasing hormone and problem behaviors in childhood. Psychoneuroendocrinology 2024; 163:106994. [PMID: 38387218 PMCID: PMC11924197 DOI: 10.1016/j.psyneuen.2024.106994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/13/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Placental corticotropin-releasing hormone (pCRH) is a neuroactive peptide produced in high concentrations in mid-late pregnancy, during key periods of fetal brain development. Some evidence suggests that higher pCRH exposure during gestation is associated with adverse neurodevelopment, particularly in female offspring. In 858 mother-child dyads from the sociodemographically diverse CANDLE cohort (Memphis, TN), we examined: (1) the slope of pCRH rise in mid-late pregnancy and (2) estimated pCRH at delivery as a measure of cumulative prenatal exposure. When children were 4 years-old, mothers reported on problem behaviors using the Child Behavior Checklist (CBCL) and cognitive performance was assessed by trained psychologists using the Stanford-Binet Intelligence Scales. We fitted linear regression models examining pCRH in relation to behavioral and cognitive performance measures, adjusting for covariates. Using interaction models, we evaluated whether associations differed by fetal sex, breastfeeding, and postnatal neighborhood opportunity. In the full cohort, log-transformed pCRH measures were not associated with outcomes; however, we observed sex differences in some models (interaction p-values≤0.01). In male offspring, an interquartile (IQR) increase in pCRH slope (but not estimated pCRH at delivery), was positively associated with raw Total (β=3.06, 95%CI: 0.40, 5.72), Internalizing (β=0.89, 95%CI: 0.03, 1.76), and Externalizing (β=1.25, 95%CI: 0.27, 2.22) Problem scores, whereas, in females, all associations were negative (Total Problems: β=-1.99, 95%CI: -3.89, -0.09; Internalizing: β=-0.82, 95%CI: -1.42, -0.23; Externalizing: β=-0.56, 95%CI: -1.34, 0.22). No associations with cognitive performance were observed nor did we observe moderation by breastfeeding or postnatal neighborhood opportunity. Our results provide further evidence that prenatal pCRH exposure may impact subsequent child behavior in sex-specific ways, however in contrast to prior studies suggesting adverse impacts in females, steeper mid-gestation pCRH rise was associated with more problem behaviors in males, but fewer in females.
Collapse
Affiliation(s)
- Emily S Barrett
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA.
| | - Alexandra Sullivan
- Center for Health and Community, University of California, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Tomomi Workman
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Yuhong Zhang
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Christine T Loftus
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Adam A Szpiro
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Alison Paquette
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - James W MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Michael Coccia
- Center for Health and Community, University of California, San Francisco, CA, USA
| | - Roger Smith
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Maria Bowman
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Alicia Smith
- Department of Gynecology and Obstetrics, Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, USA
| | - Karen Derefinko
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Ruby H N Nguyen
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - Qi Zhao
- Department of Preventive Medicine, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Sheela Sathyanarayana
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Catherine Karr
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Kaja Z LeWinn
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Nicole R Bush
- Center for Health and Community, University of California, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA; Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
23
|
Chen Y, Künzel RG, Sanchez SE, Rondon MB, Pinto NI, Sanchez E, Kirschbaum C, Valeri L, Koenen KC, Gelaye B. The Association Between Pre-Pregnancy and First-Trimester Hair Cortisol and Preterm Birth: A Causal Inference Model. RESEARCH SQUARE 2024:rs.3.rs-4095921. [PMID: 38746291 PMCID: PMC11092793 DOI: 10.21203/rs.3.rs-4095921/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Background Adverse life events and chronic psychological distress before and during pregnancy have frequently been associated with preterm birth (PTB) but the biological underpinnings remain unclear. We investigated the association between corticosteroid levels in pre-pregnancy and first-trimester hair and the risk of PTB. Methods We followed 1,808 pregnant women from a prospective pre-birth cohort study in Lima, Perú. Hair samples were taken at the end of the first pregnancy trimester. The two most proximal 3cm segments to the scalp (representing pre-pregnancy and first-trimester) were analyzed to obtain hair cortisol and cortisone concentrations (HCC and HCNC). PTB was defined as birth < 37 completed gestational weeks. We constructed four generalized propensity scores for pre-pregnancy and first-trimester HCC and HCNC to create corresponding inverse probability weights before fitting marginal structural models for estimating the effect of HCC and HCNC on PTB risk. Results Pre-pregnancy Log HCC was not independently associated with PTB risk (RR = 0.97; 95%CI: 0.79, 1.19). In contrast, one SD increase from the mean first-trimester Log HCC was independently associated with a 37% (95%CI: 1.11, 1.69) increased risk of PTB. Although imprecise, pre-pregnancy Log HCNC was negatively associated with PTB risk (RR = 0.84; 95%CI: 0.58, 1.20), whereas the association between first-trimester Log HCNC and PTB risk was positive (RR = 1.20; 95%CI: 0.87, 1.65). Conclusions Our findings show that chronic corticosteroid levels in early pregnancy are causally linked to PTB risk in pregnant Peruvian women. This finding contributes to understanding the biological underpinnings of PTB better to enhance PTB prevention.
Collapse
|
24
|
Damm J, Aureli F, Rangel-Negrín A, Barradas-Moctezuma M, Dias PAD. Analytical and biological validation of a noninvasive measurement of glucocorticoid metabolites in feces of Geoffroy's spider monkeys (Ateles geoffroyi). Am J Primatol 2024; 86:e23598. [PMID: 38258612 DOI: 10.1002/ajp.23598] [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: 08/17/2023] [Revised: 12/21/2023] [Accepted: 01/06/2024] [Indexed: 01/24/2024]
Abstract
We report on an analytical and biological validation of a commercial cortisol enzyme immunoassay (EIA) to measure glucocorticoids (GC) in feces of Geoffroy's spider monkeys (Ateles geoffroyi). Validation of endocrinological methods for each sample matrix and study species is crucial to establish that the methods produce reliable results. For the analytical validation of the EIA, we assessed parallelism, accuracy, and precision. We carried out a biological validation based on three well-studied GC patterns with the following predictions: (1) increased fecal GC metabolite (fGCM) concentrations after veterinary intervention; (2) increased fGCM concentrations during early morning hours; and (3) higher fGCM concentrations during gestation than in other female reproductive states. For the first prediction, we sampled feces of two zoo-housed females 2 days before, the day of, and 2 days after a veterinary intervention. For the second prediction, we analyzed 284 fecal samples collected from 12 wild males using a linear mixed model (LMM). For the third prediction, we analyzed 269 fecal samples of eight wild females using an LMM. Analytical validation revealed that the EIA showed parallelism, was accurate, and precise within each assay. However, there was elevated variation in between-assay precision. The biological validation supported all predictions: (1) the two zoo-housed females showed a substantial increase in fGCM concentrations 2.5 and 11 h after veterinary intervention; (2) there was a negative effect of sample collection time on fGCM concentrations (i.e., higher concentrations during early morning); (3) gestating females had significantly higher fGCM concentrations than lactating females. Thus, we analytically validated the commercial EIA and, despite between-assay variation, we were able to find three biologically relevant GC signals in captive and wild settings, and in males and females. We are therefore confident that the method can be used to noninvasively address behavioral endocrinology questions in Geoffroy's spider monkeys.
Collapse
Affiliation(s)
- Juliane Damm
- Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Mexico
| | - Filippo Aureli
- Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Mexico
- Research Centre in Evolutionary Anthropology and Palaeoecology, Liverpool John Moores University, Liverpool, UK
| | - Ariadna Rangel-Negrín
- Instituto de Neuroetología, Universidad Veracruzana, Xalapa, Mexico
- Primate Behavioral Ecology Lab, Instituto de Neuro-etología, Universidad Veracruzana, Xalapa, Mexico
| | | | - Pedro A D Dias
- Primate Behavioral Ecology Lab, Instituto de Neuro-etología, Universidad Veracruzana, Xalapa, Mexico
| |
Collapse
|
25
|
Paul N, Maiti K, Sultana Z, Fisher JJ, Zhang H, Cole N, Morgan T, Smith R. Human placenta releases extracellular vesicles carrying corticotrophin releasing hormone mRNA into the maternal blood. Placenta 2024; 146:71-78. [PMID: 38190772 DOI: 10.1016/j.placenta.2024.01.004] [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: 05/23/2023] [Revised: 12/14/2023] [Accepted: 01/03/2024] [Indexed: 01/10/2024]
Abstract
The human placenta releases diverse extracellular vesicles (EVs), including microvesicles (100-1000 nm) and exosomes (30-150 nm), into the maternal blood for feto-maternal communication. Exosomes and microvesicles contribute to normal pregnancy physiology and major pregnancy pathologies. Differences in miRNA expressions and protein content in placental exosomes have been reported in complicated pregnancies. During human pregnancy, Corticotropin-Releasing Hormone (CRH) is produced and released by the placenta into the maternal blood. CRH is involved in regulating gestational length and the initiation of labour. CRH mRNA levels in the maternal plasma rise with gestation. High levels of CRH mRNA are reported to be associated with preeclamptic and preterm pregnancies. However, the underlying mechanism of placental CRH mRNA secretion remains to be elucidated. We hypothesise that the placenta releases CRH mRNA packaged within extracellular vesicles (EVs) into the maternal blood. In this study, placental EVs (microvesicles and exosomes) were isolated from human term healthy placentas via villus washes and from explant culture media by differential centrifugation and purified by density gradient ultracentrifugation using a continuous sucrose gradient (0.25-2.5 M). Western blotting using placenta- and exosome-specific markers and electron microscopy confirmed exosomes and microvesicles in the placental wash and explant media samples. Real-time quantitative RT-PCR data detected CRH mRNA in placenta-derived EVs from placental washes and explants. We also sorted placenta-secreted EVs in maternal plasma samples (≥37 weeks) by high-resolution flow cytometry using a fluorescent-labelled PLAP antibody. CRH mRNA was demonstrated in placental EVs obtained from maternal blood plasma. We therefore show that human placental EVs carry CRH mRNA into the maternal blood. Our study implies that measuring CRH mRNA in placental EVs in the maternal plasma could beused for monitoring pregnancy.
Collapse
Affiliation(s)
- Nilanjana Paul
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, The University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia; Department of Genetic Engineering and Biotechnology, The University of Dhaka, Bangladesh
| | - Kaushik Maiti
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, The University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia
| | - Zakia Sultana
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, The University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia
| | - Joshua J Fisher
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, The University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia
| | - Huiming Zhang
- Research and Innovation Division, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Nicole Cole
- Research and Innovation Division, The University of Newcastle, University Drive, Callaghan, NSW, 2308, Australia
| | - Terry Morgan
- Department of Pathology, Oregon Health & Science University, Portland, OR, USA
| | - Roger Smith
- Mothers and Babies Research Centre, Hunter Medical Research Institute, School of Medicine and Public Health, The University of Newcastle, New Lambton Heights, New South Wales, 2305, Australia.
| |
Collapse
|
26
|
Aedla NR, Mahmood T, Ahmed B, Konje JC. Challenges in timing and mode of delivery in morbidly obese women. Best Pract Res Clin Obstet Gynaecol 2024; 92:102425. [PMID: 38150814 DOI: 10.1016/j.bpobgyn.2023.102425] [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: 09/07/2023] [Revised: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 12/29/2023]
Abstract
Globally obesity is increasing especially in the reproductive age group. Pregnant women with obesity have higher complication and intervention rates. They are also at increased risk of stillbirth and intrapartum complications. Although organisations like NICE, RCOG, ACOG and WHO have published guidelines and recommendations on care of pregnant women with obesity the evidence from which Grade A recommendations can be made on timing and how to deliver is limited. The current advice is therefore to have discussions with the woman on risks to help her make an informed decision about timing, place, and mode of delivery. Obesity is an independent risk factor for pregnancy complications including diabetes, hypertension and macrosomia. In those with these complications, the timing of delivery is often influenced by the severity of the complication. As an independent factor, population based observational studies in obese women have shown an increase in the risk of stillbirth. This risk increases linearly with weight from overweight through to class II obesity, but then rises sharply in those with class III obesity by at least 10-fold beyond 42 weeks when compared to normal weight women. This risk of stillbirth is notably higher in obese women from 34 weeks onwards compared to normal weight women. One modifiable risk factor for stillbirth as shown from various cohorts of pregnant women is prolonged pregnancy. Research has linked obesity to prolonged pregnancy. Although the exact mechanism is yet unknown some have linked this to maternal dysregulation of the hypothalamic pituitary adrenal axis leading to hormonal imbalance delaying parturition. For these women the two dilemmas are when and how best to deliver. In this review, we examine the evidence and make recommendations on the timing and mode of delivery in women with obesity. For class I obese women there are no differences in outcome with regards to timing and mode of delivery when compared to lean weight women. However, for class II and III obesity, planned induction or caesarean sections may be associated with a lower perinatal morbidity and mortality although this may be associated with an increased in maternal morbidity especially in class III obesity. Studies have shown that delivery by 39 weeks is associated with lower perinatal mortality compared to delivering after in these women. On balance the evidence would favour planned delivery (induction or caesarean section) before 40 weeks of gestation. In the morbidly obese, apart from the standard lower transverse skin incision for CS, there is evidence that a supraumbilical transverse incision may reduce morbidity but is less cosmetic. Irrespective of the option adopted, it is important to discuss the pros and cons of each.
Collapse
Affiliation(s)
- Nivedita R Aedla
- Simpsons Centre for Reproductive Medicine Royal Infirmary of Edinburgh, Edinburgh, UK.
| | | | - Badreldeen Ahmed
- Fetal Maternal Centre, Doha, Qatar; Weill Cornell Medicine Qatar. Qatar; University of Qatar, Qatar
| | - Justin C Konje
- Fetal Maternal Centre, Doha, Qatar; Weill Cornell Medicine Qatar. Qatar; Department of Health Sciences, University of Leicester, UK; University of Ho, Ghana
| |
Collapse
|
27
|
Wright MA, Giurgescu C, Misra DP, Slaughter-Acey JC, Engeland CG. Neighborhood racial composition and experiences of racial discrimination: Associations with cytokines during pregnancy among African American women. Brain Behav Immun Health 2024; 35:100715. [PMID: 38193096 PMCID: PMC10771980 DOI: 10.1016/j.bbih.2023.100715] [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: 07/19/2023] [Revised: 12/08/2023] [Accepted: 12/08/2023] [Indexed: 01/10/2024] Open
Abstract
Background Preterm birth rates are consistently higher in African American (AA) pregnancies compared to White pregnancies in the United States. Neighborhood racial composition, experiences of racial discrimination, and systemic inflammation are factors that have been associated with preterm birth and other adverse pregnancy outcomes that may account for these disparities. Here, we investigated whether perceived neighborhood racial composition and experiences of discrimination were predictive of cytokine levels during pregnancy among AA individuals. Methods 545 AA individuals completed surveys and had blood samples collected at prenatal clinics in the Midwest at three timepoints (8-18,19-29, and 30-36 weeks gestation) throughout pregnancy. Pro-inflammatory [interferon (IFN)-γ, interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, macrophage migration inhibitory factor (MIF)] and anti-inflammatory cytokines (IL-10) were quantified. Multivariate and multilevel models were used to examine associations of perceived neighborhood racial composition and experiences of racial discrimination with cytokine levels, controlling for relevant covariates. Results Perceived neighborhood racial composition was significantly associated with MIF at 30-36 weeks gestation in multivariate regression (p < 0.001). Living in neighborhoods with more compared to fewer White people was predictive of higher levels of MIF (b = 0.599, SE = 0.12, p < 0.001). Experiences of discrimination were also associated with higher levels of MIF (β = 0.141, SE = 0.07, p = 0.036). Neither predictor was associated with other cytokines. Follow-up analyses revealed that neighborhood racial composition was also predictive of higher MIF levels at 8-18 weeks gestation (p = 0.02) and at 19-29 weeks gestation (p = 0.04). Conclusions Living in neighborhoods with more White individuals and having more lifetime experiences of racial discrimination were positively related to levels of the pro-inflammatory cytokine, MIF, among pregnant AA individuals. MIF's known positive relationships with chronic stress and preterm birth suggest that these elevations in MIF may have negative health consequences. Future studies should explore whether MIF serves as a pathway between neighborhood racial composition or experiences of racial discrimination and preterm birth risk among AA individuals.
Collapse
Affiliation(s)
- Molly A. Wright
- Department of Biobehavioral Health, College of Health and Human Development, The Pennsylvania State University, University Park, PA, USA
| | - Carmen Giurgescu
- College of Nursing, University of Central Florida, Orlando, FL, USA
| | - Dawn P. Misra
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Jaime C. Slaughter-Acey
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Christopher G. Engeland
- Department of Biobehavioral Health, College of Health and Human Development, The Pennsylvania State University, University Park, PA, USA
- Ross and Carol Nese College of Nursing, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
28
|
Liu M, Ji M, Cheng J, Li Y, Tian Y, Zhao H, Wang Y, Zhu S, Zhang L, Xu X, Feng GS, Liang X, Bao H, Tang Y, Kong S, Lu J, Wang H, Lu Z, Deng W. Deciphering a critical role of uterine epithelial SHP2 in parturition initiation at single cell resolution. Nat Commun 2023; 14:7356. [PMID: 37963860 PMCID: PMC10646072 DOI: 10.1038/s41467-023-43102-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
The timely onset of female parturition is a critical determinant for pregnancy success. The highly heterogenous maternal decidua has been increasingly recognized as a vital factor in setting the timing of labor. Despite the cell type specific roles in parturition, the role of the uterine epithelium in the decidua remains poorly understood. This study uncovers the critical role of epithelial SHP2 in parturition initiation via COX1 and COX2 derived PGF2α leveraging epithelial specific Shp2 knockout mice, whose disruption contributes to delayed parturition initiation, dystocia and fetal deaths. Additionally, we also show that there are distinct types of epithelium in the decidua approaching parturition at single cell resolution accompanied with profound epithelium reformation via proliferation. Meanwhile, the epithelium maintains the microenvironment by communicating with stromal cells and macrophages. The epithelial microenvironment is maintained by a close interaction among epithelial, stromal and macrophage cells of uterine stromal cells. In brief, this study provides a previously unappreciated role of the epithelium in parturition preparation and sheds lights on the prevention of preterm birth.
Collapse
Affiliation(s)
- Meng Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Mengjun Ji
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jianghong Cheng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yingzhe Li
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yingpu Tian
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Hui Zhao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yang Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Sijing Zhu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Leilei Zhang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xinmei Xu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Gen-Sheng Feng
- Department of Pathology, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Xiaohuan Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Haili Bao
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yedong Tang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Shuangbo Kong
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jinhua Lu
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Zhongxian Lu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Wenbo Deng
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
29
|
Rinne GR, Somers JA, Ramos IF, Ross KM, Coussons-Read M, Schetter CD. Increases in maternal depressive symptoms during pregnancy and infant cortisol reactivity: Mediation by placental corticotropin-releasing hormone. Dev Psychopathol 2023; 35:1997-2010. [PMID: 35983792 PMCID: PMC9938842 DOI: 10.1017/s0954579422000621] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Maternal depressive symptoms in pregnancy may affect offspring health through prenatal programming of the hypothalamic-pituitary-adrenal (HPA) axis. The biological mechanisms that explain the associations between maternal prenatal depressive symptoms and offspring HPA axis regulation are not yet clear. This pre-registered investigation examines whether patterns of maternal depressive symptoms in pregnancy are associated with infant cortisol reactivity and whether this association is mediated by changes in placental corticotropin-releasing hormone (pCRH). METHOD A sample of 174 pregnant women completed assessments in early, mid, and late pregnancy that included standardized measures of depressive symptoms and blood samples for pCRH. Infant cortisol reactivity was assessed at 1 and 6 months of age. RESULTS Greater increases in maternal depressive symptoms in pregnancy were associated with higher cortisol infant cortisol reactivity at 1 and 6 months. Greater increases in maternal depressive symptoms in pregnancy were associated with greater increases in pCRH from early to late pregnancy which in turn were associated with higher infant cortisol reactivity. CONCLUSIONS Increases in maternal depressive symptoms and pCRH over pregnancy may contribute to higher infant cortisol reactivity. These findings help to elucidate the prenatal biopsychosocial processes contributing to offspring HPA axis regulation early in development.
Collapse
Affiliation(s)
| | | | - Isabel F. Ramos
- Department of Chicano/Latino Studies. University of California, Irvine
| | | | | | | |
Collapse
|
30
|
Arrowsmith S. Multiple pregnancies, the myometrium and the role of mechanical factors in the timing of labour. Curr Res Physiol 2023; 6:100105. [PMID: 38107788 PMCID: PMC10724211 DOI: 10.1016/j.crphys.2023.100105] [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: 05/08/2023] [Revised: 07/17/2023] [Accepted: 08/23/2023] [Indexed: 12/19/2023] Open
Abstract
Multiple pregnancy remains a relatively common occurrence, but it is associated with increased risks of adverse outcomes for the mother and her babies and presents unique challenges to healthcare providers. This review will briefly discuss multiple pregnancies, their aetiology and their problems, including preterm birth, before reviewing the processes leading to normal labour onset and how they may be different in a multiple pregnancy. The mechanisms by which mechanical factors i.e., uterine distension or 'stretch' contribute to uterine excitability and the timing of labour onset will be the major focus, and how over distention may pre-dispose multiple pregnancies to preterm birth. This includes current thinking around the role of mechano (stretch) sensitive ion channels in the myometrium and changes to other important regulators of excitability and contraction which have been identified from studies using in vitro and in vivo models of uterine stretch. Physiological stimuli arising from the fetus(es) and placenta(s) will also be discussed. In reviewing what we know about the myometrium in multiple pregnancy in humans, the focus will be on twin pregnancy as it is the most common type of multiple pregnancy and has been the most studied.
Collapse
Affiliation(s)
- Sarah Arrowsmith
- Department of Life Sciences, Manchester Metropolitan University, John Dalton Building, Chester Street, Manchester, M1 5GD, UK
| |
Collapse
|
31
|
Helegbe GK, Aryee P, Mohammed BS. Preterm Delivery and Neonatal Deaths among Anaemic Pregnant Women in the Bolgatanga Metropolis of Ghana. Anemia 2023; 2023:9865224. [PMID: 37363756 PMCID: PMC10287521 DOI: 10.1155/2023/9865224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 05/01/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Preterm deliveries and neonatal deaths as functions of anaemia in pregnancy are of major public health interest. However, data on the prevalence of preterm deliveries and their association with mortality in anaemic pregnant women in the study area are scanty. Thus, the study sought to investigate the prevalence of preterm delivery and neonatal deaths among anaemic pregnant women in the Bolgatanga Regional Hospital in the Upper East Region of Ghana during the past five years. A retrospective study design was adopted, and data were gathered between March and May 2016. Records of women who were anaemic during any trimester of their pregnancy and delivered in the hospital within the last five years were included in the study. In all, two hundred (200) cases were reviewed. Data on the sociodemographic characteristics, health status, and birth outcome of participants were captured, and analyses were conducted using SPSS version 21 while considering significant differences at p < 0.05. The study revealed that more than half of the anaemic women (52.5%, n = 105) had preterm deliveries, while neonatal mortality was 8.5% (n = 17). The proportion of mothers who received dietary or medical intervention for the treatment of anaemia and the number of attendances to antenatal clinics were comparable between preterm and normal-term mothers (p > 0.05). Mothers with preterm deliveries had a higher risk of neonatal mortality (AOR = 13.66, 95% CI = 1.65-113.30, and p=0.015). This study has shown that anaemia in pregnancy increases the risk of preterm delivery and neonatal death. It is recommended that extra care be given to pregnant women with anaemia, while further studies are conducted with a larger sample size to substantiate the claims made in this study.
Collapse
Affiliation(s)
- Gideon K. Helegbe
- Department of Biochemistry and Molecular Medicine, School of Medicine, University for Development Studies, Tamale, Ghana
| | - Paul Aryee
- Department of Nutritional Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Baba Sulemana Mohammed
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, University for Development Studies, Tamale, Ghana
| |
Collapse
|
32
|
Deer LK, Su C, Thwaites NA, Davis EP, Doom JR. A framework for testing pathways from prenatal stress-responsive hormones to cardiovascular disease risk. Front Endocrinol (Lausanne) 2023; 14:1111474. [PMID: 37223037 PMCID: PMC10200937 DOI: 10.3389/fendo.2023.1111474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/10/2023] [Indexed: 05/25/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death globally, with the prevalence projected to keep rising. Risk factors for adult CVD emerge at least as early as the prenatal period. Alterations in stress-responsive hormones in the prenatal period are hypothesized to contribute to CVD in adulthood, but little is known about relations between prenatal stress-responsive hormones and early precursors of CVD, such as cardiometabolic risk and health behaviors. The current review presents a theoretical model of the relation between prenatal stress-responsive hormones and adult CVD through cardiometabolic risk markers (e.g., rapid catch-up growth, high BMI/adiposity, high blood pressure, and altered blood glucose, lipids, and metabolic hormones) and health behaviors (e.g., substance use, poor sleep, poor diet and eating behaviors, and low physical activity levels). Emerging evidence in human and non-human animal literatures suggest that altered stress-responsive hormones during gestation predict higher cardiometabolic risk and poorer health behaviors in offspring. This review additionally highlights limitations of the current literature (e.g., lack of racial/ethnic diversity, lack of examination of sex differences), and discusses future directions for this promising area of research.
Collapse
Affiliation(s)
- LillyBelle K. Deer
- Department of Psychology, University of Denver, Denver, CO, United States
| | - Chen Su
- Department of Psychology, University of Denver, Denver, CO, United States
| | | | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, United States
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA, United States
| | - Jenalee R. Doom
- Department of Psychology, University of Denver, Denver, CO, United States
| |
Collapse
|
33
|
Gallo DM, Romero R, Bosco M, Gotsch F, Jaiman S, Jung E, Suksai M, Ramón Y Cajal CL, Yoon BH, Chaiworapongsa T. Meconium-stained amniotic fluid. Am J Obstet Gynecol 2023; 228:S1158-S1178. [PMID: 37012128 PMCID: PMC10291742 DOI: 10.1016/j.ajog.2022.11.1283] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 04/04/2023]
Abstract
Green-stained amniotic fluid, often referred to as meconium-stained amniotic fluid, is present in 5% to 20% of patients in labor and is considered an obstetric hazard. The condition has been attributed to the passage of fetal colonic content (meconium), intraamniotic bleeding with the presence of heme catabolic products, or both. The frequency of green-stained amniotic fluid increases as a function of gestational age, reaching approximately 27% in post-term gestation. Green-stained amniotic fluid during labor has been associated with fetal acidemia (umbilical artery pH <7.00), neonatal respiratory distress, and seizures as well as cerebral palsy. Hypoxia is widely considered a mechanism responsible for fetal defecation and meconium-stained amniotic fluid; however, most fetuses with meconium-stained amniotic fluid do not have fetal acidemia. Intraamniotic infection/inflammation has emerged as an important factor in meconium-stained amniotic fluid in term and preterm gestations, as patients with these conditions have a higher rate of clinical chorioamnionitis and neonatal sepsis. The precise mechanisms linking intraamniotic inflammation to green-stained amniotic fluid have not been determined, but the effects of oxidative stress in heme catabolism have been implicated. Two randomized clinical trials suggest that antibiotic administration decreases the rate of clinical chorioamnionitis in patients with meconium-stained amniotic fluid. A serious complication of meconium-stained amniotic fluid is meconium aspiration syndrome. This condition develops in 5% of cases presenting with meconium-stained amniotic fluid and is a severe complication typical of term newborns. Meconium aspiration syndrome is attributed to the mechanical and chemical effects of aspirated meconium coupled with local and systemic fetal inflammation. Routine naso/oropharyngeal suctioning and tracheal intubation in cases of meconium-stained amniotic fluid have not been shown to be beneficial and are no longer recommended in obstetrical practice. A systematic review of randomized controlled trials suggested that amnioinfusion may decrease the rate of meconium aspiration syndrome. Histologic examination of the fetal membranes for meconium has been invoked in medical legal litigation to time the occurrence of fetal injury. However, inferences have been largely based on the results of in vitro experiments, and extrapolation of such findings to the clinical setting warrants caution. Fetal defecation throughout gestation appears to be a physiologic phenomenon based on ultrasound as well as in observations in animals.
Collapse
Affiliation(s)
- 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, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI; Department of Gynecology and Obstetrics, Universidad Del Valle, Cali, Colombia
| | - 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, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI; Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI.
| | - Mariachiara Bosco
- 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, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Francesca Gotsch
- 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, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Sunil Jaiman
- 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, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Pathology, Wayne State University School of Medicine, Detroit, MI
| | - 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, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - 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, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Carlos López Ramón Y Cajal
- Unit of Prenatal Diagnosis, Service of Obstetrics and Gynecology, Álvaro Cunqueiro Hospital, Vigo, Spain
| | - Bo Hyun Yoon
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - 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, United States Department of Health and Human Services, Bethesda, MD, and Detroit, MI; Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| |
Collapse
|
34
|
Monitoring uterine contractions during labor: current challenges and future directions. Am J Obstet Gynecol 2023; 228:S1192-S1208. [PMID: 37164493 DOI: 10.1016/j.ajog.2022.10.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/22/2022] [Accepted: 10/27/2022] [Indexed: 03/21/2023]
Abstract
Organ-level models are used to describe how cellular and tissue-level contractions coalesce into clinically observable uterine contractions. More importantly, these models provide a framework for evaluating the many different contraction patterns observed in laboring patients, ideally offering insight into the pitfalls of currently available recording modalities and suggesting new directions for improving recording and interpretation of uterine contractions. Early models proposed wave-like propagation of bioelectrical activity as the sole mechanism for recruiting the myometrium to participate in the contraction and increase contraction strength. However, as these models were tested, the results consistently revealed that sequentially propagating waves do not travel long distances and do not encompass the gravid uterus. To resolve this discrepancy, a model using 2 mechanisms, or a "dual model," for organ-level signaling has been proposed. In the dual model, the myometrium is recruited by action potentials that propagate wave-like as far as 10 cm. At longer distances, the myometrium is recruited by a mechanotransduction mechanism that is triggered by rising intrauterine pressure. In this review, we present the influential models of uterine function, highlighting their main features and inconsistencies, and detail the role of intrauterine pressure in signaling and cervical dilation. Clinical correlations demonstrate the application of organ-level models. The potential to improve the recording and clinical interpretation of uterine contractions when evaluating labor is discussed, with emphasis on uterine electromyography. Finally, 7 questions are posed to help guide future investigations on organ-level signaling mechanisms.
Collapse
|
35
|
Lee JH, Torpy DJ. Adrenal insufficiency in pregnancy: Physiology, diagnosis, management and areas for future research. Rev Endocr Metab Disord 2023; 24:57-69. [PMID: 35816262 DOI: 10.1007/s11154-022-09745-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 02/01/2023]
Abstract
Adrenal insufficiency requires prompt diagnosis in pregnancy, as untreated, it can lead to serious consequences such as adrenal crisis, intrauterine growth restriction and even foetal demise. Similarities between symptoms of adrenal insufficiency and those of normal pregnancy can complicate diagnosis. Previously diagnosed adrenal insufficiency needs monitoring and, often, adjustment of adrenal hormone replacement. Many physiological changes occur to the hypothalamic-pituitary-adrenal (HPA) axis during pregnancy, often making diagnosis and management of adrenal insufficiency challenging. Pregnancy is a state of sustained physiologic hypercortisolaemia; there are multiple contributing factors including high plasma concentrations of placental derived corticotropin-releasing hormone (CRH), adrenocorticotropin (ACTH) and increased adrenal responsiveness to ACTH. Despite increased circulating concentrations of CRH-binding protein (CRH-BP) and the major cortisol binding protein, corticosteroid binding globulin (CBG), free concentrations of both hormones are increased progressively in pregnancy. In addition, pregnancy leads to activation of the renin-angiotensin-aldosterone system. Most adrenocortical hormone diagnostic thresholds are not applicable or validated in pregnancy. The management of adrenal insufficiency also needs to reflect the physiologic changes of pregnancy, often requiring increased doses of glucocorticoid and at times mineralocorticoid replacement, especially in the last trimester. In this review, we describe pregnancy induced changes in adrenal function, the diagnosis and management of adrenal insufficiency in pregnancy and areas requiring further research.
Collapse
Affiliation(s)
- Jessica H Lee
- Department of Medicine, University of Adelaide, Adelaide, South Australia, Australia.
| | - David J Torpy
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia.
| |
Collapse
|
36
|
Gaspari L, Soyer-Gobillard MO, Rincheval N, Paris F, Kalfa N, Hamamah S, Sultan C. Birth Outcomes in DES Children and Grandchildren: A Multigenerational National Cohort Study on Informative Families. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2542. [PMID: 36767903 PMCID: PMC9915936 DOI: 10.3390/ijerph20032542] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 06/18/2023]
Abstract
OBJECTIVE Diethylstilbestrol (DES), a potent synthetic nonsteroidal estrogen belonging to the family of endocrine disrupting chemicals (EDCs), can cross the placenta and may cause permanent adverse health effects in the exposed mothers, their children (exposed in utero), and also their grandchildren through germline contribution to the zygote. This study evaluated pregnancy duration and birthweight (BW) variations in the children and grandchildren born before, during, and after maternal DES treatment in the same informative families, to rule out genetic, endocrine, and environmental factors. DESIGN AND SETTING Nationwide retrospective observational study on 529 families of DES-treated women registered at the HHORAGES-France Association. The inclusion criteria were: (i) women with at least three pregnancies and three viable children among whom the first was not exposed in utero to DES, followed by one or more children with fetal exposure to DES, and then by one or more children born after DES treatment; (ii) women with at least one pre-DES or post-DES grandchild and one DES grandchild; (iii) confirmed data on total DES dose. Women with severe pathologies or whose illness status, habitat, lifestyle habits, profession, treatment changed between pregnancies, and all mothers who reported pregnancy-related problems, were excluded. RESULTS In all, 74 women met all criteria. The preterm birth (PTB) rate was 2.7% in pre-DES, 14.9% in DES, and 10.8% in post-DES children (Cochran-Armitage test for trend, p = 0.0095). The mean BW was higher in DES than pre-DES full-term neonates (≥37 weeks of gestation) (p = 0.007). In grandchildren, BW was not different, whereas the PTB and low BW rates were slightly increased in children of DES women. CONCLUSIONS These data within the same informative families show the DES impact on BW and PTB in DES and post-DES children and grandchildren. In particular, mean BW was higher in DES than pre-DES full-term neonates. This result may be in opposition to previous data from American cohorts, which reported lower BW in DES children, but is consistent with animal study. Our retrospective observational study highlights a multigenerational and likely transgenerational effect of this EDC in humans.
Collapse
Affiliation(s)
- Laura Gaspari
- CHU Montpellier, Université de Montpellier, Unité d’Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie, 34295 Montpellier, France
- CHU Montpellier, Université de Montpellier, Centre de Référence Maladies Rares du Développement Génital, Constitutif Sud, Hôpital Lapeyronie, 34295 Montpellier, France
- Université de Montpellier, INSERM 1203, DEFE, 34295 Montpellier, France
| | | | - Nathalie Rincheval
- Equipe de Recherche AESIO SANTE, Clinique Beausoleil, 34070 Montpellier, France
| | - Françoise Paris
- CHU Montpellier, Université de Montpellier, Unité d’Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie, 34295 Montpellier, France
- CHU Montpellier, Université de Montpellier, Centre de Référence Maladies Rares du Développement Génital, Constitutif Sud, Hôpital Lapeyronie, 34295 Montpellier, France
- Université de Montpellier, INSERM 1203, DEFE, 34295 Montpellier, France
| | - Nicolas Kalfa
- CHU Montpellier, Université de Montpellier, Centre de Référence Maladies Rares du Développement Génital, Constitutif Sud, Hôpital Lapeyronie, 34295 Montpellier, France
- CHU Montpellier, Université de Montpellier, Département de Chirurgie Pédiatrique, Hôpital Lapeyronie, 34295 Montpellier, France
| | - Samir Hamamah
- Université de Montpellier, INSERM 1203, DEFE, 34295 Montpellier, France
- CHU Montpellier, Université de Montpellier, Service de Biologie de la Reproduction, 34295 Montpellier, France
| | - Charles Sultan
- CHU Montpellier, Université de Montpellier, Unité d’Endocrinologie-Gynécologie Pédiatrique, Service de Pédiatrie, 34295 Montpellier, France
| |
Collapse
|
37
|
Kyathanahalli C, Snedden M, Hirsch E. Is human labor at term an inflammatory condition?†. Biol Reprod 2023; 108:23-40. [PMID: 36173900 PMCID: PMC10060716 DOI: 10.1093/biolre/ioac182] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 01/20/2023] Open
Abstract
Parturition at term in normal pregnancy follows a predictable sequence of events. There is some evidence that a state of inflammation prevails in the reproductive tissues during labor at term, but it is uncertain whether this phenomenon is the initiating signal for parturition. The absence of a clear temporal sequence of inflammatory events prior to labor casts doubt on the concept that normal human labor at term is primarily the result of an inflammatory cascade. This review examines evidence linking parturition and inflammation in order to address whether inflammation is a cause of labor, a consequence of labor, or a separate but related phenomenon. Finally, we identify and suggest ways to reconcile inconsistencies regarding definitions of labor onset in published research, which may contribute to the variability in conclusions regarding the genesis and maintenance of parturition. A more thorough understanding of the processes underlying normal parturition at term may lead to novel insights regarding abnormal labor, including spontaneous preterm labor, preterm premature rupture of the fetal membranes, and dysfunctional labor, and the role of inflammation in each.
Collapse
Affiliation(s)
- Chandrashekara Kyathanahalli
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Madeline Snedden
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Emmet Hirsch
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
38
|
Watt M, Mohammadzadeh P, Pinsinski E, Hollinshead FK, Bouma GJ. Corticotropin releasing hormone is present in the feline placenta and maternal serum. Front Endocrinol (Lausanne) 2023; 14:1132743. [PMID: 37124751 PMCID: PMC10140359 DOI: 10.3389/fendo.2023.1132743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/22/2023] [Indexed: 05/02/2023] Open
Abstract
Background In women, placental corticotropin releasing hormone (CRH) can be detected in maternal blood throughout pregnancy and is important in the regulation of the timing of parturition. However, its role in other mammalian species is unclear. In fact, very little is known about the presence and localization of CRH in placentas other than human. In this study we report for the first time the presence of CRH in feline placenta and maternal serum. Methods Presence of CRH mRNA and protein was assessed using RT-PCR and Western blot, respectively, in at term domestic cat placentas opportunistically obtained at a local animal shelter and spay clinic. In addition, CRH localization within the placenta was demonstrated via immunohistochemistry. Finally, presence of CRH in maternal blood from early (¾21 days) and mid (25-35 days) stages of pregnancy was investigated by ELISA. Results CRH mRNA and protein were detected in feline placentas, and localized to larger decidual cells and fetal trophoblast cells, including the binucleate cells. CRH was detectable in maternal blood collected from early-stage pregnancies, and amounts significantly increased in mid-gestation samples. Conclusion This is the first report on the presence and localization of CRH in the feline placenta, and its increase in maternal serum during the first half of pregnancy. These data lay the foundation for future studies to determine if CRH can be used as potential novel marker for early pregnancy diagnosis, determination, and monitoring in felids, and could greatly increase efficiency and success in zoo breeding programs utilizing artificial reproductive technologies for endangered feline species.
Collapse
Affiliation(s)
- Madison Watt
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Pardis Mohammadzadeh
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Emma Pinsinski
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Fiona K. Hollinshead
- Department of Clinical Sciences, Colorado State University, Fort Collins, CO, United States
- *Correspondence: Fiona K. Hollinshead,
| | - Gerrit J. Bouma
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
39
|
Paquette AG, MacDonald J, Bammler T, Day DB, Loftus CT, Buth E, Mason WA, Bush NR, Lewinn KZ, Marsit C, Litch JA, Gravett M, Enquobahrie DA, Sathyanarayana S. Placental transcriptomic signatures of spontaneous preterm birth. Am J Obstet Gynecol 2023; 228:73.e1-73.e18. [PMID: 35868418 PMCID: PMC9790028 DOI: 10.1016/j.ajog.2022.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 07/01/2022] [Accepted: 07/09/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Spontaneous preterm birth accounts for most preterm births and leads to significant morbidity in the newborn and childhood period. This subtype of preterm birth represents an increasing proportion of all preterm births when compared with medically indicated preterm birth, yet it is understudied in omics analyses. The placenta is a key regulator of fetal and newborn health, and the placental transcriptome can provide insight into pathologic changes that lead to spontaneous preterm birth. OBJECTIVE This analysis aimed to identify genes for which placental expression was associated with spontaneous preterm birth (including early preterm and late preterm birth). STUDY DESIGN The ECHO PATHWAYS consortium extracted RNA from placental samples collected from the Conditions Affecting Neurocognitive Development and Learning in Early Childhood and the Global Alliance to Prevent Prematurity and Stillbirth studies. Placental transcriptomic data were obtained by RNA sequencing. Linear models were fit to estimate differences in placental gene expression between term birth and spontaneous preterm birth (including gestational age subgroups defined by the American College of Obstetricians and Gynecologists). Models were adjusted for numerous confounding variables, including labor status, cohort, and RNA sequencing batch. This analysis excluded patients with induced labor, chorioamnionitis, multifetal gestations, or medical indications for preterm birth. Our combined cohort contained gene expression data for 14,023 genes in 48 preterm and 540 term samples. Genes and pathways were considered statistically significantly different at false discovery rate-adjusted P value of <.05. RESULTS In total, we identified 1728 genes for which placental expression was associated with spontaneous preterm birth with more differences in expression in early preterm samples than late preterm samples when compared with full-term samples. Of those, 9 genes were significantly decreased in both early and late spontaneous preterm birth, and the strongest associations involved placental expression of IL1B, ALPL, and CRLF1. In early and late preterm samples, we observed decreased expression of genes involved in immune signaling, signal transduction, and endocrine function. CONCLUSION This study provides a comprehensive assessment of the differences in the placental transcriptome associated with spontaneous preterm birth with robust adjustment for confounding. Results of this study are in alignment with the known etiology of spontaneous preterm birth, because we identified multiple genes and pathways for which the placental and chorioamniotic membrane expression was previously associated with prematurity, including IL1B. We identified decreased expression in key signaling pathways that are essential for placental growth and function, which may be related to the etiology of spontaneous preterm birth. We identified increased expression of genes within metabolic pathways associated exclusively with early preterm birth. These signaling and metabolic pathways may provide clinically targetable pathways and biomarkers. The findings presented here can be used to understand underlying pathologic changes in premature placentas, which can inform and improve clinical obstetrics practice.
Collapse
Affiliation(s)
- Alison G Paquette
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA; Department of Pediatrics, University of Washington, Seattle, WA.
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Theo Bammler
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Drew B Day
- Center for Child Health, Behavior, and Development, Seattle Children's Research Institute, Seattle, WA
| | - Christine T Loftus
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - Erin Buth
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA
| | - W Alex Mason
- Department of Preventative Medicine, University of Tennessee Health Science Center, Memphis, TN
| | - Nicole R Bush
- Department of Psychiatry and Behavioral Sciences, University of San Francisco, San Francisco, CA; Department of Pediatrics, University of San Francisco, San Francisco, CA
| | - Kaja Z Lewinn
- Department of Psychiatry and Behavioral Sciences, University of San Francisco, San Francisco, CA
| | - Carmen Marsit
- Gangarosa Department of Environmental Health, Emory University, Atlanta, GA
| | - James A Litch
- Global Alliance to Prevent Preterm Birth and Stillbirth (GAPPS), Lynnwood, WA
| | - Michael Gravett
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA
| | | | - Sheela Sathyanarayana
- Department of Pediatrics, University of Washington, Seattle, WA; Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA; Center for Child Health, Behavior, and Development, Seattle Children's Research Institute, Seattle, WA; Department of Epidemiology, University of Washington, Seattle, WA
| |
Collapse
|
40
|
Herrera CL, Maiti K, Smith R. Preterm Birth and Corticotrophin-Releasing Hormone as a Placental Clock. Endocrinology 2022; 164:bqac206. [PMID: 36478045 PMCID: PMC10583728 DOI: 10.1210/endocr/bqac206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Preterm birth worldwide remains a significant cause of neonatal morbidity and mortality, yet the exact mechanisms of preterm parturition remain unclear. Preterm birth is not a single condition, but rather a syndrome with a multifactorial etiology. This multifactorial nature explains why individual predictive measures for preterm birth have had limited sensitivity and specificity. One proposed pathway for preterm birth is via placentally synthesized corticotrophin-releasing hormone (CRH). CRH is a peptide hormone that increases exponentially in pregnancy and has been implicated in preterm birth because of its endocrine, autocrine, and paracrine roles. CRH has actions that increase placental production of estriol and of the transcription factor nuclear factor-κB, that likely play a key role in activating the myometrium. CRH has been proposed as part of a placental clock, with early activation of placental production resulting in preterm birth. This article will review the current understanding of preterm birth, CRH as an initiator of human parturition, and the evidence regarding the use of CRH in the prediction of preterm birth.
Collapse
Affiliation(s)
- Christina L Herrera
- Department of Obstetrics & Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9032, USA
| | - Kaushik Maiti
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales 2305, Australia
| | - Roger Smith
- Mothers and Babies Research Centre, Hunter Medical Research Institute, University of Newcastle, Newcastle, New South Wales 2305, Australia
| |
Collapse
|
41
|
Corticotropin-Releasing Hormone: Biology and Therapeutic Opportunities. BIOLOGY 2022; 11:biology11121785. [PMID: 36552294 PMCID: PMC9775501 DOI: 10.3390/biology11121785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/16/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
In 1981, Wylie Vale, Joachim Spiess, Catherine Rivier, and Jean Rivier reported on the characterization of a 41-amino-acid peptide from ovine hypothalamic extracts with high potency and intrinsic activity stimulating the secretion of adrenocorticotropic hormone and β-endorphin by cultured anterior pituitary cells. With its sequence known, this neuropeptide was determined to be a hormone and consequently named corticotropin-releasing hormone (CRH), although the term corticotropin-releasing factor (CRF) is still used and preferred in some circumstances. Several decades have passed since this seminal contribution that opened a new research era, expanding the understanding of the coding of stress-related processes. The characterization of CRH receptors, the availability of CRH agonists and antagonists, and advanced immunocytochemical staining techniques have provided evidence that CRH plays a role in the regulation of several biological systems. The purpose of this review is to summarize the present knowledge of this 41-amino-acid peptide.
Collapse
|
42
|
Alterations in maternal sFlt-1 and PlGF: Time to labor onset in term-/late-term pregnancies with and without placental dysfunction. Pregnancy Hypertens 2022; 30:148-153. [DOI: 10.1016/j.preghy.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 11/07/2022]
|
43
|
Clark J, Bulka CM, Martin CL, Roell K, Santos HP, O'Shea TM, Smeester L, Fry R, Dhingra R. Placental epigenetic gestational aging in relation to maternal sociodemographic factors and smoking among infants born extremely preterm: a descriptive study. Epigenetics 2022; 17:2389-2403. [PMID: 36134874 PMCID: PMC9665142 DOI: 10.1080/15592294.2022.2125717] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 11/03/2022] Open
Abstract
Social determinants of health (SDoH) are defined as the conditions in which people are born, grow, live, work, and age. The distribution of these conditions is influenced by underlying structural factors and may be linked to adverse pregnancy outcomes through epigenetic modifications of gestational tissues. A promising modification is epigenetic gestational age (eGA), which captures 'biological' age at birth. Measuring eGA in placenta, an organ critical for foetal development, may provide information about how SDoH 'get under the skin' during pregnancy to influence birth outcomes and ethnic/racial disparities. We examined relationships of placental eGA with sociodemographic factors, smoking, and two key clinical outcomes: Apgar scores and NICU length of stay. Using the Robust Placental Clock, we estimated eGA for placental samples from the Extremely Low Gestational Age Newborns cohort (N = 408). Regression modelling revealed smoking during pregnancy was associated with placental eGA acceleration (i.e., eGA higher than chronologic gestational age). This association differed by maternal race: among infants born to mothers racialized as Black, we observed greater eGA acceleration (+0.89 week, 95% CI: 0.38, 1.40) as compared to those racialized as white (+0.27 week, 95% CI: -0.06, 0.59). Placental eGA acceleration was also correlated with shorter NICU lengths of stay, but only among infants born to mothers racialized as Black (-0.08 d/week-eGA, 95% CI: -0.12, -0.05). Together, these observed associations suggest that interpretations of epigenetic gestational aging may be tissue-specific.
Collapse
Affiliation(s)
- Jeliyah Clark
- Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Catherine M Bulka
- Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Chantel L Martin
- Department of Epidemiology, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Kyle Roell
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC, USA
| | - Hudson P Santos
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC, USA
- Biobehavioral Lab, School of Nursing, University of North Carolina, Chapel Hill, North Carolina, USA
| | - T Michael O'Shea
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lisa Smeester
- Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC, USA
| | - Rebecca Fry
- Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC, USA
- Curriculum in Toxicology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Radhika Dhingra
- Department of Environmental Sciences and Engineering, UNC Gillings School of Global Public Health, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
44
|
MAYNE GB, DeWITT PE, RINGHAM B, WARRENER AG, CHRISTIANS U, DABELEA D, HURT KJ. A Nested Case-Control Study of Allopregnanolone and Preterm Birth in the Healthy Start Cohort. J Endocr Soc 2022; 7:bvac179. [PMID: 36632210 PMCID: PMC9825133 DOI: 10.1210/jendso/bvac179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Indexed: 11/26/2022] Open
Abstract
Context Chronic stress is a risk factor for preterm birth; however, objective measures of stress in pregnancy are limited. Maternal stress biomarkers may fill this gap. Steroid hormones and neurosteroids such as allopregnanolone (ALLO) play important roles in stress physiology and pregnancy maintenance and therefore may be promising for preterm birth prediction. Objective We evaluated maternal serum ALLO, progesterone, cortisol, cortisone, pregnanolone, and epipregnanolone twice in gestation to evaluate associations with preterm birth. Methods We performed a nested case-control study using biobanked fasting serum samples from the Healthy Start prebirth cohort. We included healthy women with a singleton pregnancy and matched preterm cases with term controls (1:1; N = 27 per group). We used a new HPLC-tandem mass spectrometry assay to quantify ALLO and five related steroids. We used ANOVA, Fisher exact, χ2, t test, and linear and logistic regression as statistical tests. Results Maternal serum ALLO did not associate with preterm birth nor differ between groups. Mean cortisol levels were significantly higher in the preterm group early in pregnancy (13w0d-18w0d; P < 0.05) and higher early pregnancy cortisol associated with increased odds of preterm birth (at 13w0d; odds ratio, 1.007; 95% CI, 1.0002-1.014). Progesterone, cortisone, pregnanolone, and epipregnanolone did not associate with preterm birth. Conclusion The findings from our pilot study suggest potential utility of cortisol as a maternal serum biomarker for preterm birth risk assessment in early pregnancy. Further evaluation using larger cohorts and additional gestational timepoints for ALLO and the other analytes may be informative.
Collapse
Affiliation(s)
- Gabriella B MAYNE
- Department of Anthropology, University of Colorado, Denver, CO 80204, USA
| | - Peter E DeWITT
- Department of Pediatrics Informatics and Data Science, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Brandy RINGHAM
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Anna G WARRENER
- Department of Anthropology, University of Colorado, Denver, CO 80204, USA
| | - Uwe CHRISTIANS
- iC42 Clinical Research & Development, Department of Anesthesiology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Dana DABELEA
- Lifecourse Epidemiology of Adiposity and Diabetes Center, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - K Joseph HURT
- Correspondence: K. Joseph Hurt, MD, PhD, 12700 East 19th Ave, Aurora, CO 80045, USA.
| |
Collapse
|
45
|
Oaks BM, Adu-Afarwuah S, Ashorn P, Lartey A, Laugero KD, Okronipa H, Stewart CP, Dewey KG. Increased risk of preterm delivery with high cortisol during pregnancy is modified by fetal sex: a cohort study. BMC Pregnancy Childbirth 2022; 22:727. [PMID: 36151538 PMCID: PMC9502964 DOI: 10.1186/s12884-022-05061-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background Previous studies show an association between maternal plasma and salivary cortisol and preterm birth but have been primarily conducted in high-income countries. It is unknown whether salivary cortisol is a risk factor for preterm birth in Ghana. Our objective was to determine whether maternal salivary cortisol during pregnancy was associated with pregnancy duration and preterm delivery in Ghana. Methods We conducted a cohort study of 783 pregnant women in Ghana. We measured salivary cortisol at baseline (mean 16 wk), 28 wk., and 36 wk. gestation. Pregnancy duration was determined primarily by ultrasound. We used adjusted linear regression models to examine the association between cortisol and pregnancy duration and Poisson regression models to determine the risk of preterm delivery among women with high cortisol at baseline or 28 wk. gestation. Results Mean pregnancy duration was 39.4 ± 1.8 wk. and 6.6% had a preterm delivery. Mean maternal cortisol increased throughout pregnancy, from 4.9 ± 2.7 nmol/L at baseline (16 wk) to 6.4 ± 3.2 nmol/L at 28 wk. and 7.9 ± 3.0 nmol/L at 36 wk. gestation. In adjusted analyses, higher cortisol concentrations at baseline (β = − 0.39, p = .002) and 28 wk. (β = − 0.49, p = .001), but not 36 wk. (β = − 0.23, p = .084) were associated with a shorter pregnancy duration. Women with high cortisol at baseline (> 6.3 nmol/L) had an increased relative risk of preterm delivery (RR (95% CI): 1.96 (1.13, 3.40)), but the association between high cortisol at 28 wk. and preterm delivery was not significant. There was a significant interaction with fetal sex (p-for-interaction = 0.037): among women carrying male fetuses, high cortisol at baseline increased the risk of preterm delivery threefold (3.18 (1.51, 6.71)) while there was no association (1.17 (0.50, 2.74)) among women carrying female fetuses. Conclusion Higher maternal cortisol is associated with a shorter pregnancy duration and an increased risk of preterm delivery. Subgroup analysis by fetal sex revealed that this association is evident primarily among women carrying male fetuses. Future studies of cortisol and preterm delivery should include consideration of fetal sex as a potential effect modifier. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-05061-8.
Collapse
Affiliation(s)
- Brietta M Oaks
- Department of Nutrition and Food Sciences, University of Rhode Island, Kingston, RI, 02881, USA.
| | - Seth Adu-Afarwuah
- Department of Nutrition and Food Science, University of Ghana, Accra, Ghana
| | - Per Ashorn
- Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health, Technology, Tampere University, Tampere, Finland.,Department of Pediatrics, Tampere University Hospital, Tampere, Finland
| | - Anna Lartey
- Department of Nutrition and Food Science, University of Ghana, Accra, Ghana
| | - Kevin D Laugero
- USDA Western Human Nutrition Research Center, Davis, CA, USA.,Department of Nutrition, University of California, Davis, CA, USA
| | - Harriet Okronipa
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK, USA
| | | | - Kathryn G Dewey
- Department of Nutrition, University of California, Davis, CA, USA
| |
Collapse
|
46
|
Characterization of CRH-Binding Protein (CRHBP) in Chickens: Molecular Cloning, Tissue Distribution and Investigation of Its Role as a Negative Feedback Regulator within the Hypothalamus–Pituitary–Adrenal Axis. Genes (Basel) 2022; 13:genes13101680. [PMID: 36292565 PMCID: PMC9601729 DOI: 10.3390/genes13101680] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 01/08/2023] Open
Abstract
Corticotropin (ACTH) is a pituitary hormone playing important roles in stress response within the hypothalamus–pituitary–adrenal (HPA) axis. The biosynthesis and secretion of ACTH are controlled by multiple factors, including corticotropin-releasing hormone (CRH). As a key hypothalamus-derived regulator, CRH binds to corticotropin-releasing hormone receptor 1 (CRHR1) in the anterior pituitary gland to regulate ACTH synthesis and release. Thus, CRH-binding protein (CRHBP), which binds CRH with high affinity to inhibit CRH-induced ACTH secretion from pituitary cells, draws wide attention. In contrast to the extensive investigation of CRHBP in mammals and other lower vertebrates, the gene structure, tissue expression and physiological functions of CRHBP in birds remain largely unknown. In the present study, using chicken (c-) as our animal model, we examined the gene structure, tissue expression and functionality of CRHBP. Our results showed that: (1) cCRHBP cDNA encodes a 345 amino acid precursor, which shares high sequence identity with that of mammals, reptiles, frogs and fish; (2) cCRHBP is abundantly expressed in the brain (cerebrum and hypothalamus), pituitary and ovary; (3) cCRHBP inhibits the signaling of cCRHRs induced by cCRH, thus reducing the cCRH-induced ACTH secretion from cultured chick pituitary cells; (4) stress mediators (e.g., glucocorticoids) and stress significantly upregulate CRHBP mRNA expression in chickens, supporting its role as a negative feedback regulator in the HPA axis. The present study enriches our understanding of the conserved roles of CRHBP across vertebrates. In addition, chicken is an important poultry animal with multiple economic traits which are tightly controlled by the HPA axis. The characterization of the chicken CRHBP gene helps to reveal the molecular basis of the chicken HPA axis and is thus beneficial to the poultry industry.
Collapse
|
47
|
Özdemir BG, Özdemir H, Atalay CR. The importance of fetal adrenal gland volume measurement in successful labor induction with oxytocin. J Obstet Gynaecol Res 2022; 48:2514-2521. [PMID: 35817550 DOI: 10.1111/jog.15361] [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: 12/13/2021] [Revised: 02/23/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022]
Abstract
AIM We aimed to show the predictive value of fetal adrenal gland volume (FAGV) measurement in the success of labor induction with oxytocin. METHODS This is a prospective cohort study that included a total of 103 term nulliparous. Immediately after deciding to induce labor with oxytocin, the fetal right and left adrenal gland measurements were obtained. RESULTS Induction success was achieved in 89 of 103 (86.4%) pregnant women. In comparison of the FAGV values of failed induction labor group, the total volumes of right and left fetal adrenal glands and the fetal zone volumes (FZV) were found to be statistically significantly higher in the successful labor induction group (p < 0.001). In predicting induction success, the cut-off value of fetal adrenal gland volume (cFAGV) was set as >230.2 mm3 /kg and area under curve (AUC) value of 0.872 (95% confidence interval [CI], 0.736-1.000) for right total adrenal gland (p < 0.001). The cut-off value of cFAGV was set as >236.7 mm3 /kg and AUC value of 0.891(95%CI, 0.768-1.000) (p < 0.001) for left total adrenal gland. The cut-off value of cFAGV was set as >12.6 mm3 /kg and AUC value of 0.952 (95%CI, 0.905-0.999) for right FZV(p < 0.001). The cut-off value of cFAGV was set as >7.8 mm3 /kg and AUC value of 0.884 (95%CI, 0.752-1.000) for left FZV (p < 0.001). CONCLUSIONS Two-dimension ultrasonographic measurement is an easy-to-access and noninvasive method that can be integrated into the algorithms to predict the success of induction based on FAGV measurement.
Collapse
Affiliation(s)
- Belma G Özdemir
- Republic of Turkey Ministry of Health Ankara City Hospital, Obstetrics and Gynecology Department, Ankara, Turkey
| | - Halis Özdemir
- Malatya Training and Research Hospital, Department of Obstetrics and Gynecology, Perinatology Clinic, Malatya, Turkey
| | - Cemal R Atalay
- Republic of Turkey Ministry of Health Ankara City Hospital, Obstetrics and Gynecology Department, Ankara, Turkey
| |
Collapse
|
48
|
Cervantes O, Talavera IC, Every E, Coler B, Li M, Li A, Li H, Adams Waldorf K. Role of hormones in the pregnancy and sex-specific outcomes to infections with respiratory viruses. Immunol Rev 2022; 308:123-148. [PMID: 35373371 PMCID: PMC9189035 DOI: 10.1111/imr.13078] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/13/2023]
Abstract
Pregnant women infected with pathogenic respiratory viruses, such as influenza A viruses (IAV) and coronaviruses, are at higher risk for mortality, hospitalization, preterm birth, and stillbirth. Several factors are likely to contribute to the susceptibility of pregnant individuals to severe lung disease including changes in pulmonary physiology, immune defenses, and effector functions of some immune cells. Pregnancy is also a physiologic state characterized by higher levels of multiple hormones that may impact the effector functions of immune cells, such as progesterone, estrogen, human chorionic gonadotropin, prolactin, and relaxin. Each of these hormones acts to support a tolerogenic immune state of pregnancy, which helps prevent fetal rejection, but may also contribute to an impaired antiviral response. In this review, we address the unique role of adaptive and innate immune cells in the control of pathogenic respiratory viruses and how pregnancy and specific hormones can impact their effector actions. We highlight viruses with sex-specific differences in infection outcomes and why pregnancy hormones may contribute to fetal protection but aid the virus at the expense of the mother's health.
Collapse
Affiliation(s)
- Orlando Cervantes
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Irene Cruz Talavera
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Emma Every
- University of Washington School of Medicine, Spokane, Washington, United States of America
| | - Brahm Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, United States of America
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Department of Biological Sciences, Columbia University, New York City, New York, United States of America
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Case Western Reserve, Cleveland, Ohio, United States of America
| | - Hanning Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
49
|
Ramos IF, Ross KM, Rinne GR, Somers JA, Mancuso RA, Hobel CJ, Coussons-Read M, Dunkel Schetter C. Pregnancy anxiety, placental corticotropin-releasing hormone and length of gestation. Biol Psychol 2022; 172:108376. [PMID: 35667479 PMCID: PMC10022399 DOI: 10.1016/j.biopsycho.2022.108376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 05/29/2022] [Accepted: 05/31/2022] [Indexed: 12/20/2022]
Abstract
OBJECTIVE High pregnancy anxiety is a consistent predictor of earlier labor and delivery. Placental corticotropin-releasing hormone (pCRH) predicts earlier delivery consistently and it has been identified as a biological mediator of the association between pregnancy anxiety and gestational length. However, studies have not examined whether changes in pregnancy anxiety are associated with earlier birth as mediated by changes in pCRH during pregnancy. Accordingly, this study tests whether linear changes in pregnancy anxiety are associated with length of gestation indirectly through nonlinear increases in pCRH over pregnancy. METHODS A sample of pregnant women (n=233) completed prenatal assessments in early pregnancy, second trimester, and third trimester that included a 4-item assessment of pregnancy anxiety and collection of blood samples assayed for pCRH using radioimmunoassay. Length of gestation was abstracted from medical records after birth. RESULTS Increases in pregnancy anxiety from early pregnancy to third trimester predicted shorted length of gestation, as did nonlinear increases in pCRH over pregnancy. However, there was no evidence of an indirect effect of changes in pregnancy anxiety on length of gestation via changes in pCRH. CONCLUSIONS These results indicate that linear changes in pregnancy anxiety and nonlinear changes in pCRH during pregnancy are independent risk factors for shortened gestational length. This study adds to a small but growing body of work on biopsychological processes in pregnancy and length of gestation. Modeling changes in psychological and biological processes during pregnancy could provide more insight into understanding risk for adverse pregnancy outcomes.
Collapse
|
50
|
Kawka-Paciorkowska K, Bręborowicz GH. The role of corticoliberin concentration levels and placental CRH receptors 1 and 2 in the prolongation of pregnancy. Gynecol Endocrinol 2022; 38:478-482. [PMID: 35475389 DOI: 10.1080/09513590.2022.2068521] [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] [Indexed: 10/18/2022] Open
Abstract
OBJECTIVE To examine the changes in CRH concentrations in the blood serum of pregnant women and in the placenta of patients after the 41st week of gestation, and to determine its influence on the effectiveness of inducing labor and its progress. MATERIALS AND METHODS The study group comprised pregnant patients who did not deliver until the 41 week of gestation (n = 114). The control group was divided into two subgroups: patients in whom delivery started spontaneously before the 41st week of gestation (n = 24) and pregnant patients in whom delivery started spontaneously after the 41st week of gestation (n = 23). Blood serum and placenta were obtained from the patients. Corticoliberin originating from blood serum was assessed with the use of ELISA Kit. Parts of the placenta were stained with monoclonal antibodies for the presence of corticoliberin, corticoliberin receptors 1 and 2. RESULTS No statistically significant differences were found with regard to corticoliberin concentrations in the blood or during a qualitative assessment of the number of CRH R1 in the placenta between the research groups. However, corticoliberin receptor 2 had a statistically higher expression rate in the control group in which the delivery started spontaneously before the 41st week of gestation. CONCLUSION In post-term pregnancy, the up-regulation of CRH R2 receptor is disturbed with no change in CRH R1 expression, which complicates the initiation of labor despite correct corticoliberin levels in both blood serum and the placenta. Pregnancy duration over 41 weeks and the effectiveness of preinducing or inducing labor do not depend on corticoliberin concentrations.
Collapse
|