1
|
Nonn O, Debnath O, Valdes DS, Sallinger K, Secener AK, Fischer C, Tiesmeyer S, Nimo J, Kuenzer T, Ulrich J, Maxian T, Knöfler M, Karau P, Bartolomaeus H, Kroneis T, Frolova A, Neuper L, Haase N, Malt A, Müller-Bötticher N, Kräker K, Kedziora S, Forstner D, Eils R, Schmidt-Ullrich R, Haider S, Verlohren S, Stern C, Sugulle M, Jones S, Thilaganathan B, Kaitu’u-Lino TJ, Tong S, Huppertz B, El-Heliebi A, Staff AC, Coscia F, Müller DN, Dechend R, Gauster M, Ishaque N, Herse F. Senescent Syncytiotrophoblast Secretion During Early Onset Preeclampsia. Hypertension 2025; 82:787-799. [PMID: 39440423 PMCID: PMC12002046 DOI: 10.1161/hypertensionaha.124.23362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Preeclampsia is a severe hypertensive disorder in pregnancy that causes preterm delivery, maternal and fetal morbidity, mortality, and life-long sequelae. Understanding the pathogenesis of preeclampsia is a critical first step toward protecting mother and child from this syndrome and increased risk of cardiovascular disease later in life. However, effective early predictive tests and therapies for preeclampsia are scarce. METHODS To identify novel markers and signaling pathways for early onset preeclampsia, we profiled human maternal-fetal interface units (fetal villi and maternal decidua) from early onset preeclampsia and healthy controls using single-nucleus RNA sequencing combined with spatial transcriptomics. The placental syncytiotrophoblast is in direct contact with maternal blood and forms the barrier between fetal and maternal circulation. RESULTS We identified different transcriptomic states of the endocrine syncytiotrophoblast nuclei with patterns of dysregulation associated with a senescence-associated secretory phenotype and a spatial dysregulation of senescence in the placental trophoblast layer. Elevated senescence markers were validated in placental tissues of clinical multicenter cohorts. Importantly, several secreted senescence-associated secretory phenotype factors were elevated in maternal blood already in the first trimester. We verified the secreted senescence markers, PAI-1 (plasminogen activator inhibitor 1) and activin A, as identified in our single-nucleus RNA sequencing model as predictive markers before clinical preeclampsia diagnosis. CONCLUSIONS This indicates that increased syncytiotrophoblast senescence appears weeks before clinical manifestation of early onset preeclampsia, suggesting that the dysregulated preeclamptic placenta starts with higher cell maturation resulting in premature and increased senescence-associated secretory phenotype release. These senescence-associated secretory phenotype markers may serve as an additional early diagnostic tool for this syndrome.
Collapse
Affiliation(s)
- Olivia Nonn
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Olivia Debnath
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Daniela S. Valdes
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Katja Sallinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
- Centre for Biomarker Research in Medicine, Graz, Austria (K.S., T. Kroneis, A.E.-H.)
| | - Ali Kerim Secener
- Max Delbrück Center - Berlin Institute for Medical Systems Biology (MDC-BIMSB), Berlin, Germany (A.K.S., C.F.)
- Department of Biology, Chemistry and Pharmacy, Institute of Chemistry and Biochemistry, Freie Universität Berlin, Germany (A.K.S.)
| | - Cornelius Fischer
- Max Delbrück Center - Berlin Institute for Medical Systems Biology (MDC-BIMSB), Berlin, Germany (A.K.S., C.F.)
| | - Sebastian Tiesmeyer
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Jose Nimo
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| | - Thomas Kuenzer
- Institute for Medical Informatics, Statistics and Documentation (T. Kuenzer), Medical University of Graz, Austria
| | - Juliane Ulrich
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| | - Theresa Maxian
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Austria (T.M., M.K., S.H.)
| | - Martin Knöfler
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Austria (T.M., M.K., S.H.)
| | - Philipp Karau
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Hendrik Bartolomaeus
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Thomas Kroneis
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
- Centre for Biomarker Research in Medicine, Graz, Austria (K.S., T. Kroneis, A.E.-H.)
| | - Alina Frolova
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- Institute of Molecular Biology and Genetic of the National Academy of Sciences of Ukraine (NASU), Kyiv, Ukraine (A.F.)
| | - Lena Neuper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Nadine Haase
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Alexander Malt
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Niklas Müller-Bötticher
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Kristin Kräker
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Sarah Kedziora
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Roland Eils
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Ruth Schmidt-Ullrich
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| | - Sandra Haider
- Department of Obstetrics and Gynaecology, Reproductive Biology Unit, Medical University of Vienna, Austria (T.M., M.K., S.H.)
| | - Stefan Verlohren
- Department of Obstetrics and Gynaecology, Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (S.V.)
| | - Christina Stern
- Department of Obstetrics and Gynaecology, University Hospital Graz, Medical University Graz, Austria (C.S.)
| | - Meryam Sugulle
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway (M.S., A.C.S.)
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (M.S., A.C.S.)
| | - Stuart Jones
- Clinical Biochemistry, King George’s Hospital, London, United Kingdom (S.J.)
| | - Basky Thilaganathan
- Fetal Medicine Unit, St George’s University Hospitals NHS Foundation Trust, London, United Kingdom (B.T.)
| | - Tu’uhevaha J. Kaitu’u-Lino
- Department of Obstetrics and Gynaecology (Mercy Hospital for Women), The University of Melbourne, VIC, Australia (T.J.K.-L., S. Tong)
| | - Stephen Tong
- Department of Obstetrics and Gynaecology (Mercy Hospital for Women), The University of Melbourne, VIC, Australia (T.J.K.-L., S. Tong)
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Amin El-Heliebi
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
- Centre for Biomarker Research in Medicine, Graz, Austria (K.S., T. Kroneis, A.E.-H.)
| | - Anne Cathrine Staff
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway (M.S., A.C.S.)
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Norway (M.S., A.C.S.)
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| | - Dominik N. Müller
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
| | - Ralf Dechend
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
- German Center for Cardiovascular Research (DZHK), Berlin, Germany (O.N., D.S.V., H.B., N.H., K.K., S.K., D.N.M., R.D.)
- Department of Cardiology and Nephrology, HELIOS Clinic, Berlin, Germany (R.D.)
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center (O.N., K.S., T. Kroneis, L.N., D.F., B.H., A.E.-H., M.G.), Medical University of Graz, Austria
| | - Naveed Ishaque
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Center of Digital Health, Germany (O.D., S. Tiesmeyer, P.K., A.M., N.M.-B., R.E., N.I.)
| | - Florian Herse
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., D.N.M., R.D., F.H.)
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité – Universitätsmedizin Berlin, Germany (O.N., D.S.V., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., D.N.M., R.D., F.H.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (O.N., D.S.V., J.N., J.U., H.B., A.F., N.H., K.K., S.K., R.S.-U., F.C., D.N.M., R.D., F.H.)
| |
Collapse
|
2
|
Botha SM, Bartho LA, Hartmann S, Cannon P, Nguyen A, Nguyen TV, Pritchard N, Dechend R, Nonn O, Tong S, Kaitu'u-Lino TJ. Cystatin 6 (CST6) and Legumain (LGMN) are potential mediators in the pathogenesis of preeclampsia. Sci Rep 2025; 15:12945. [PMID: 40234537 PMCID: PMC12000359 DOI: 10.1038/s41598-025-96823-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
Preeclampsia results from placental insufficiency and causes maternal endothelial dysfunction and multi-organ damage. Our in-silico analysis identified Cystatin 6 (CST6), a cysteine protease inhibitor, as located on the placental surface where it might be released into maternal circulation. This study aimed to characterise CST6 and one of its high affinity targets, Legumain (LGMN), in preeclampsia and assess its biomarker potential by measuring levels in maternal circulation. Placental CST6 mRNA expression was significantly increased in 78 pregnancies complicated by early-onset preeclampsia (delivering at < 34 weeks' gestation) relative to 30 gestation matched controls (P < 0.0001). LGMN mRNA expression was significantly decreased (P = 0.0309). Circulating CST6 was increased in 35 pregnancies complicated by early-onset preeclampsia (< 34 weeks' gestation) relative to 27 gestation matched controls (P = 0.0261), and LGMN levels remained unchanged. At 36 weeks' gestation, circulating CST6 was significantly increased (P = 0.001), while LGMN was significantly decreased (P = 0.0135) in 21 pregnancies preceding diagnosis of preeclampsia at term, compared to 184 pregnancies that did not develop preeclampsia. Human trophoblast stem cells (hTSC) were differentiated into syncytiotrophoblast or extravillous trophoblast (EVT) to evaluate CST6 and LGMN expression in these trophoblast lineages. CST6 and LGMN mRNA expression were significantly increased across 96 h after syncytiotrophoblast (P = 0.0066 and P = 0.0010 respectively) and EVT differentiation (P = 0.0618 and P = 0.0016 respectively), with the highest expression in syncytiotrophoblast. Computational analysis of two publicly available single-cell and single-nuclei RNA sequencing datasets correlated with the expression pattern observed in vitro. When syncytiotrophoblast cells were exposed to hypoxia (1% O2 vs. 8% O2), CST6 expression significantly increased (P = 0.0079), whilst LGMN expression was unchanged. The vascular endothelium may serve as an additional source of circulating CST6 and LGMN in preeclampsia. Induction of dysfunction in endothelial cells by TNFα, caused reduced CST6 expression (P = 0.0036), whilst LGMN expression remained unchanged. Administering recombinant CST6 to endothelial cells enhanced markers of endothelial dysfunction and LGMN expression in the presence of TNFα. These findings indicate an inverse relationship between CST6 and LGMN in the placenta and maternal circulation in preeclampsia. We suggest elevated circulating levels of CST6 may be induced by placental hypoxia. This study provides novel insight into the dysregulation of CST6 and LGMN in preeclampsia and introduces their potential roles in human pregnancy and associated pathology.
Collapse
Affiliation(s)
- Stefan M Botha
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia.
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- Experimental and Clinical Research Center, a cooperation between Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany.
| | - Lucy A Bartho
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Sunhild Hartmann
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Ping Cannon
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Anna Nguyen
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tuong-Vi Nguyen
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Natasha Pritchard
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Ralf Dechend
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Department of Cardiology and Nephrology, HELIOS Clinic, Berlin, Germany
| | - Olivia Nonn
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation between Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Stephen Tong
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, The Department of Obstetrics, Gynaecology and Newborn Health, Mercy Hospital for Women, University of Melbourne, Heidelberg, Victoria, 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
3
|
Bartho LA, Walker SP, Cannon P, Nguyen TV, Nguyen A, Botha SM, Hannan NJ, Tong S, Kaitu'u-Lino TJ. Leukocyte-associated immunoglobulin-like receptor 1 (LAIR1) is reduced with preeclampsia and small for gestational aged fetuses. Placenta 2024; 156:10-13. [PMID: 39216164 DOI: 10.1016/j.placenta.2024.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/11/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Leukocyte-associated immunoglobulin-like receptor 1 (LAIR1) is an inhibitory receptor expressed on immune cells. We evaluated LAIR1 in placentas from preeclamptic or small for gestational age (SGA) pregnancies, and placental explant model (1 % O2, IL6 and TNFα, or control). LAIR1 mRNA was reduced in placentas from preeclamptic (p < 0.0001, n = 78) and SGA (p < 0.0001, n = 32) pregnancies. LAIR1 protein expression was reduced in placentas from preeclampsia (p < 0.0001, n = 43) and SGA (p = 0.009, n = 10) pregnancies. Hypoxia (1 % O2) reduced LAIR1 mRNA expression in placental explants (p = 0.008). These findings suggest hypoxia modulates LAIR1 expression in the placenta.
Collapse
Affiliation(s)
- Lucy A Bartho
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia.
| | - Susan P Walker
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Tuong-Vi Nguyen
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Anna Nguyen
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Stefan M Botha
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität at Berlin and Humboldt-Universität, Berlin, Germany; Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center. for Molecular Medicine in the Helmholtz Association and the Charité - Universitätsmedizin Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; DZHK (German Center for Cardiovascular Research), partner site, Berlin, Germany
| | - Natalie J Hannan
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| |
Collapse
|
4
|
Gibbins KJ, Roberts VHJ, Lo JO, Boniface ER, Schabel MC, Silver RM, Frias AE. MRI assessed placental volume and adverse pregnancy outcomes: Secondary analysis of prospective cohort study. Placenta 2024; 154:168-175. [PMID: 39018609 PMCID: PMC11368624 DOI: 10.1016/j.placenta.2024.07.007] [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: 02/01/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
INTRODUCTION Our goal was to evaluate the potential utility of magnetic resonance imaging (MRI) placental volume as an assessment of placental insufficiency. METHODS Secondary analysis of a prospective cohort undergoing serial placental MRIs at two academic tertiary care centers. The population included 316 participants undergoing MRI up to three times throughout gestation. MRI was used to calculate placental volume in milliliters (ml). Placental-mediated adverse pregnancy outcome (cAPO) included preeclampsia with severe features, abnormal antenatal surveillance, and perinatal mortality. Serial measurements were grouped as time point 1 (TP1) <22 weeks, TP2 22 0/7-29 6/7 weeks, and TP3 ≥30 weeks. Mixed effects models compared change in placental volume across gestation between cAPO groups. Association between cAPO and placental volume was determined using logistic regression at each TP with discrimination evaluated using area under receiver operator curve (AUC). Placental volume was then added to known clinical predictive variables and evaluated with test characteristics and calibration. RESULTS 59 (18.7 %) of 316 participants developed cAPO. Placental volume growth across gestation was slower in the cAPO group (p < 0.001). Placental volume was lower in the cAPO group at all time points, and alone was moderately predictive of cAPO at TP3 (AUC 0.756). Adding placental volume to clinical variables had moderate discrimination at all time points, with strongest test characteristics at TP3 (AUC 0.792) with sensitivity of 77.5 % and specificity of 75.3 % at a predicted probability cutoff of 15 %. DISCUSSION MRI placental volume warrants further study for assessment of placental insufficiency, particularly later in gestation.
Collapse
Affiliation(s)
- Karen J Gibbins
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon, USA.
| | - Victoria H J Roberts
- Oregon National Primate Research Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Jamie O Lo
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Oregon National Primate Research Center, Portland, Oregon, USA
| | - Emily R Boniface
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon, USA
| | - Matthias C Schabel
- Advanced Imaging Resource Center, Oregon Health & Science University, Portland, Oregon, USA
| | - Robert M Silver
- Department of Obstetrics & Gynecology, University of Utah Health, Salt Lake City, Utah, USA
| | - Antonio E Frias
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
5
|
Juusela A, Jung E, Gallo DM, Bosco M, Suksai M, Diaz-Primera R, Tarca AL, Than NG, Gotsch F, Romero R, Tinnakorn Chaiworapongsa. Maternal plasma syndecan-1: a biomarker for fetal growth restriction. J Matern Fetal Neonatal Med 2023; 36:2150074. [PMID: 36597808 PMCID: PMC10291740 DOI: 10.1080/14767058.2022.2150074] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 11/14/2022] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The identification of fetal growth disorders is an important clinical priority given that they increase the risk of perinatal morbidity and mortality as well as long-term diseases. A subset of small-for-gestational-age (SGA) infants are growth-restricted, and this condition is often attributed to placental insufficiency. Syndecan-1, a product of the degradation of the endothelial glycocalyx, has been proposed as a biomarker of endothelial damage in different pathologies. During pregnancy, a "specialized" form of the glycocalyx-the "syncytiotrophoblast glycocalyx"-covers the placental villi. The purpose of this study was to determine whether the concentration of maternal plasma syndecan-1 can be proposed as a biomarker for fetal growth restriction. STUDY DESIGN A cross-sectional study was designed to include women with normal pregnancy (n = 130) and pregnant women who delivered an SGA neonate (n = 50). Doppler velocimetry of the uterine and umbilical arteries was performed in women with an SGA fetus at the time of diagnosis. Venipuncture was performed within 48 h of Doppler velocimetry and plasma concentrations of syndecan-1 were determined by a specific and sensitive immunoassay. RESULTS (1) Plasma syndecan-1 concentration followed a nonlinear increase with gestational age in uncomplicated pregnancies (R2 = 0.27, p < .001); (2) women with a pregnancy complicated with an SGA fetus had a significantly lower mean plasma concentration of syndecan-1 than those with an appropriate-for-gestational-age fetus (p = .0001); (3) this difference can be attributed to fetal growth restriction, as the mean plasma syndecan-1 concentration was significantly lower only in the group of women with an SGA fetus who had abnormal umbilical and uterine artery Doppler velocimetry compared to controls (p = .00071; adjusted p = .0028). A trend toward lower syndecan-1 concentrations was also noted for SGA with abnormal uterine but normal umbilical artery Doppler velocimetry (p = .0505; adjusted p = .067); 4) among women with an SGA fetus, those with abnormal umbilical and uterine artery Doppler findings had a lower mean plasma syndecan-1 concentration than women with normal Doppler velocimetry (p = .02; adjusted p = .04); 5) an inverse relationship was found between the maternal plasma syndecan-1 concentration and the umbilical artery pulsatility index (r = -0.5; p = .003); and 6) a plasma syndecan-1 concentration ≤ 850 ng/mL had a positive likelihood ratio of 4.4 and a negative likelihood ratio of 0.24 for the identification of a mother with an SGA fetus who had abnormal umbilical artery Doppler velocimetry (area under the ROC curve 0.83; p < .001). CONCLUSION Low maternal plasma syndecan-1 may reflect placental diseases and this protein could be a biomarker for fetal growth restriction. However, as a sole biomarker for this condition, its accuracy is low.
Collapse
Affiliation(s)
- Alexander Juusela
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Eunjung Jung
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Dahiana M. Gallo
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Mariachiara Bosco
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Manaphat Suksai
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Ramiro Diaz-Primera
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Adi L. Tarca
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Computer Science, Wayne State University College of Engineering, Detroit, Michigan, USA
| | - Nandor Gabor Than
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Systems Biology of Reproduction Research Group, Institute of Enzymology, Research Centre for Natural Sciences, Budapest, Hungary
- Maternity Private Clinic, Budapest, Hungary
| | - Francesca Gotsch
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, United States Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
6
|
Hu M, Zhang Y, Zhang X, Zhang X, Huang X, Lu Y, Li Y, Brännström M, Sferruzzi-Perri AN, Shao LR, Billig H. Defective Uterine Spiral Artery Remodeling and Placental Senescence in a Pregnant Rat Model of Polycystic Ovary Syndrome. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1916-1935. [PMID: 37689383 DOI: 10.1016/j.ajpath.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/28/2023] [Accepted: 08/23/2023] [Indexed: 09/11/2023]
Abstract
Pregnancy-related problems have been linked to impairments in maternal uterine spiral artery (SpA) remodeling. The mechanisms underlying this association are still unclear. It is also unclear whether hyperandrogenism and insulin resistance, the two common manifestations of polycystic ovary syndrome, affect uterine SpA remodeling. We verified previous work in which exposure to 5-dihydrotestosterone (DHT) and insulin (INS) in rats during pregnancy resulted in hyperandrogenism, insulin intolerance, and higher fetal mortality. Exposure to DHT and INS dysregulated the expression of angiogenesis-related genes in the uterus and placenta and also decreased expression of endothelial nitric oxide synthase and matrix metallopeptidases 2 and 9, increased fibrotic collagen deposits in the uterus, and reduced expression of marker genes for SpA-associated trophoblast giant cells. These changes were related to a greater proportion of unremodeled uterine SpAs and a smaller proportion of highly remodeled arteries in DHT + INS-exposed rats. Placentas from DHT + INS-exposed rats exhibited decreased basal and labyrinth zone regions, reduced maternal blood spaces, diminished labyrinth vascularity, and an imbalance in the abundance of vascular and smooth muscle proteins. Furthermore, placentas from DHT + INS-exposed rats showed expression of placental insufficiency markers and a significant increase in cell senescence-associated protein levels. Altogether, this work demonstrates that increased pregnancy complications in polycystic ovary syndrome may be mediated by problems with uterine SpA remodeling, placental functionality, and placental senescence.
Collapse
Affiliation(s)
- Min Hu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China; Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yuehui Zhang
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xu Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - XiuYing Zhang
- Department of Obstetrics and Gynecology, Key Laboratory and Unit of Infertility in Chinese Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyue Huang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yaxing Lu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Yijia Li
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China; Institute of Integrated Traditional Chinese Medicine and Western Medicine, Guangzhou Medical University, Guangzhou, China
| | - Mats Brännström
- Department of Obstetrics and Gynecology, Sahlgrenska University Hospital, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Linus R Shao
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Håkan Billig
- Department of Physiology/Endocrinology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
7
|
Zhang Y, Lai J, Wang X, Li M, Zhang Y, Ji C, Chen Q, Lu S. Genome-wide single nucleotide polymorphism (SNP) data reveal potential candidate genes for litter traits in a Yorkshire pig population. Arch Anim Breed 2023; 66:357-368. [PMID: 38111388 PMCID: PMC10726026 DOI: 10.5194/aab-66-357-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/10/2023] [Indexed: 12/20/2023] Open
Abstract
The litter trait is one of the most important economic traits, and increasing litter size is of great economic value in the pig industry. However, the molecular mechanisms underlying pig litter traits remain elusive. To identify molecular markers and candidate genes for pig litter traits, a genome-wide association study (GWAS) and selection signature analysis were conducted in a Yorkshire pig population. A total of 518 producing sows were genotyped with Illumina Porcine SNP 50 BeadChip, and 1969 farrowing records for the total number born (TNB), the number born alive (NBA), piglets born dead (PBD), and litter weight born alive (LWB) were collected. Then, a GWAS was performed for the four litter traits using a repeatability model. Based on the estimated breeding values (EBVs) of TNB, 15 high- and 15 low-prolificacy individuals were selected from the 518 sows to implement selection signature analysis. Subsequently, the selection signatures affecting the litter traits of sows were detected by using two methods including the fixation index (FST) and θ π . Combining the results of the GWAS and selection signature analysis, 20 promising candidate genes (NKAIN2, IGF1R, KISS1R, TYRO3, SPINT1, ADGRF5, APC2, PTBP1, CLCN3, CBR4, HPF1, FAM174A, SCP2, CLIC1, ZFYVE9, SPATA33, KIF5C, EPC2, GABRA2, and GABRA4) were identified. These findings provide novel insights into the genetic basis of pig litter traits and will be helpful for improving the reproductive performances of sows in pig breeding.
Collapse
Affiliation(s)
- Yu Zhang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Jinhua Lai
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Xiaoyi Wang
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Mingli Li
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yanlin Zhang
- Yunnan Fuyuefa Livestock and Poultry Feeding Company Limited, Kunming, 650300, China
| | - Chunlv Ji
- Yunnan Fuyuefa Livestock and Poultry Feeding Company Limited, Kunming, 650300, China
| | - Qiang Chen
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Shaoxiong Lu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| |
Collapse
|
8
|
Chen L, Li J, You Y, Qian Z, Liu J, Jiang Y, Gu Y, Xiao J, Zhang Y. Secreted proteins in plasma and placenta as novel non-invasive biomarkers for intrahepatic cholestasis of pregnancy: A case-control study. Heliyon 2023; 9:e21616. [PMID: 38027820 PMCID: PMC10661505 DOI: 10.1016/j.heliyon.2023.e21616] [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: 07/06/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background Intrahepatic cholestasis of pregnancy (ICP) is likely to lead to unfavorable consequences. Total bile acid (TBA) is thought to be the sole ICP indicator available as of now, but it comes with some kind of restrictions in terms of sensitivity and specificity. We were endeavoring to find potential diagnostic biomarkers for ICP in this investigation. Methods This case-control study with a prospective nature included 40 females in the stage of pregnancy who were diagnosed with ICP. It also included another 20 females who were also pregnant but with sound physical condition(control). Placental and plasma samples were collected from all females that were in the stage of pregnancy, except for 20 ICP patients, in which only plasma was collected. We used four-dimensional data-independent acquisition followed by enzyme-linked immunosorbent assay and immunohistochemistry to identify and validate plasma and placental profiles in ICP patients and controls. Bioinformatics was adopted in an effort to demonstrate the relevant biological processes and signalling pathways. Correlation analysis was used to analyse the consistency of tissue and plasma protein expression and the correlation between sequencing and experimental results. Results The expression levels of nectin-1 (NECTIN1), Kunitz-type protease inhibitor 1 (SPINT1), and inter-alpha-trypsin inhibitor heavy chain H3 (ITIH3) were remarkably higher in ICP patients than in controls. However, heparin cofactor 2 (SERPIND1) expression levels in female participants in the stage of pregnancy who were diagnosed with ICP were remarkably lower than those pregnant females with good physical fitness. In addition to the negative correlation between SERPIND1 and TBA, NECTIN1, SPINT1, and ITIH3 expression positively correlated with TBA. Area under the receiver operating characteristic curve (AUC) values of 0.7925, 0.8313, 0.8163, and 0.9025, respectively, were used to assess the diagnostic accuracies of NECTIN1, SPINT1, ITIH3, and SERPIND1. AUC (0.9438) was considerably greater when NECTIN1, SPINT1, and SERPIND1 were integrated, according to binary logistic regression. The AUC of the ROC curve for various combinations of SERPIND1 and other indicators was higher than itself, thus providing a more reliable ICP diagnosis. Furthermore, according to the bioinformatics analysis, the NECTIN1, SPINT1, ITIH3, and SERPIND1 were identified as secreted proteins because they were localized in the extracellular region. Conclusions This research discovered new non-invasive ICP indicators. On top of this, it sheds new light on the crucial diagnostic function of secreted proteins in ICP.
Collapse
Affiliation(s)
- Lingyan Chen
- Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Jingyang Li
- Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Yilan You
- Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Zhiwen Qian
- Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Jiayu Liu
- Wuxi Maternal and Child Health Care Hospital, Jiangnan University, Wuxi, 214002, China
| | - Ying Jiang
- Wuxi Maternal and Child Health Care Hospital, Jiangnan University, Wuxi, 214002, China
| | - Ying Gu
- Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Jianping Xiao
- Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
| | - Yan Zhang
- Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, 214002, China
- Wuxi Maternal and Child Health Care Hospital, Jiangnan University, Wuxi, 214002, China
| |
Collapse
|
9
|
Miranda J, Paules C, Noell G, Youssef L, Paternina-Caicedo A, Crovetto F, Cañellas N, Garcia-Martín ML, Amigó N, Eixarch E, Faner R, Figueras F, Simões RV, Crispi F, Gratacós E. Similarity network fusion to identify phenotypes of small-for-gestational-age fetuses. iScience 2023; 26:107620. [PMID: 37694157 PMCID: PMC10485038 DOI: 10.1016/j.isci.2023.107620] [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: 01/05/2023] [Revised: 04/19/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Fetal growth restriction (FGR) affects 5-10% of pregnancies, is the largest contributor to fetal death, and can have long-term consequences for the child. Implementation of a standard clinical classification system is hampered by the multiphenotypic spectrum of small fetuses with substantial differences in perinatal risks. Machine learning and multiomics data can potentially revolutionize clinical decision-making in FGR by identifying new phenotypes. Herein, we describe a cluster analysis of FGR based on an unbiased machine-learning method. Our results confirm the existence of two subtypes of human FGR with distinct molecular and clinical features based on multiomic analysis. In addition, we demonstrated that clusters generated by machine learning significantly outperform single data subtype analysis and biologically support the current clinical classification in predicting adverse maternal and neonatal outcomes. Our approach can aid in the refinement of clinical classification systems for FGR supported by molecular and clinical signatures.
Collapse
Affiliation(s)
- Jezid Miranda
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universidad de Cartagena, Cartagena de Indias, Colombia
| | - Cristina Paules
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Aragon Institute of Health Research (IIS Aragon), Obstetrics Department, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | - Guillaume Noell
- University of Barcelona, Biomedicine Department, IDIBAPS, Centre for Biomedical Research on Respiratory Diseases (CIBERES), Barcelona, Spain
| | - Lina Youssef
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | | | - Francesca Crovetto
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Nicolau Cañellas
- Metabolomics Platform, IISPV, DEEiA, Universidad Rovira i Virgili, Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Tarragona, Spain
| | - María L. Garcia-Martín
- BIONAND, Andalusian Centre for Nanomedicine and Biotechnology, Junta de Andalucía, Universidad de Málaga, Málaga, Spain
| | | | - Elisenda Eixarch
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rosa Faner
- University of Barcelona, Biomedicine Department, IDIBAPS, Centre for Biomedical Research on Respiratory Diseases (CIBERES), Barcelona, Spain
| | - Francesc Figueras
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Rui V. Simões
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
- Institute for Research & Innovation in Health (i3S), University of Porto, Porto, Portugal
| | - Fàtima Crispi
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| | - Eduard Gratacós
- BCNatal – Barcelona Center for Maternal-Fetal and Neonatal Medicine (Hospital Clínic and Hospital Sant Joan de Deu), IDIBAPS, University of Barcelona, and Centre for Biomedical Research on Rare Diseases (CIBER-ER), Barcelona, Spain
| |
Collapse
|
10
|
Espinosa CA, Khan W, Khanam R, Das S, Khalid J, Pervin J, Kasaro MP, Contrepois K, Chang AL, Phongpreecha T, Michael B, Ellenberger M, Mehmood U, Hotwani A, Nizar A, Kabir F, Wong RJ, Becker M, Berson E, Culos A, De Francesco D, Mataraso S, Ravindra N, Thuraiappah M, Xenochristou M, Stelzer IA, Marić I, Dutta A, Raqib R, Ahmed S, Rahman S, Hasan ASMT, Ali SM, Juma MH, Rahman M, Aktar S, Deb S, Price JT, Wise PH, Winn VD, Druzin ML, Gibbs RS, Darmstadt GL, Murray JC, Stringer JSA, Gaudilliere B, Snyder MP, Angst MS, Rahman A, Baqui AH, Jehan F, Nisar MI, Vwalika B, Sazawal S, Shaw GM, Stevenson DK, Aghaeepour N. Multiomic signals associated with maternal epidemiological factors contributing to preterm birth in low- and middle-income countries. SCIENCE ADVANCES 2023; 9:eade7692. [PMID: 37224249 PMCID: PMC10208584 DOI: 10.1126/sciadv.ade7692] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 04/20/2023] [Indexed: 05/26/2023]
Abstract
Preterm birth (PTB) is the leading cause of death in children under five, yet comprehensive studies are hindered by its multiple complex etiologies. Epidemiological associations between PTB and maternal characteristics have been previously described. This work used multiomic profiling and multivariate modeling to investigate the biological signatures of these characteristics. Maternal covariates were collected during pregnancy from 13,841 pregnant women across five sites. Plasma samples from 231 participants were analyzed to generate proteomic, metabolomic, and lipidomic datasets. Machine learning models showed robust performance for the prediction of PTB (AUROC = 0.70), time-to-delivery (r = 0.65), maternal age (r = 0.59), gravidity (r = 0.56), and BMI (r = 0.81). Time-to-delivery biological correlates included fetal-associated proteins (e.g., ALPP, AFP, and PGF) and immune proteins (e.g., PD-L1, CCL28, and LIFR). Maternal age negatively correlated with collagen COL9A1, gravidity with endothelial NOS and inflammatory chemokine CXCL13, and BMI with leptin and structural protein FABP4. These results provide an integrated view of epidemiological factors associated with PTB and identify biological signatures of clinical covariates affecting this disease.
Collapse
Affiliation(s)
- Camilo A. Espinosa
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Waqasuddin Khan
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Rasheda Khanam
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sayan Das
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Javairia Khalid
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Jesmin Pervin
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Margaret P. Kasaro
- University of North Carolina Global Projects Zambia, Lusaka, Zambia
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Alan L. Chang
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Thanaphong Phongpreecha
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Basil Michael
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Mathew Ellenberger
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Usma Mehmood
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Aneeta Hotwani
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Ambreen Nizar
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Furqan Kabir
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Ronald J. Wong
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin Becker
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Eloise Berson
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony Culos
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Davide De Francesco
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Samson Mataraso
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Neal Ravindra
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Melan Thuraiappah
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Maria Xenochristou
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Ina A. Stelzer
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Ivana Marić
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Arup Dutta
- Centre for Public Health Kinetics, New Delhi, Delhi, India
| | - Rubhana Raqib
- International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | | | | | | | - Said M. Ali
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Mohamed H. Juma
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Monjur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Shaki Aktar
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Saikat Deb
- Centre for Public Health Kinetics, New Delhi, Delhi, India
- Public Health Laboratory—Ivo de Carneri, Pemba, Zanzibar, Tanzania
| | - Joan T. Price
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Obstetrics and Gynaecology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Paul H. Wise
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Virginia D. Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maurice L. Druzin
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ronald S. Gibbs
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Gary L. Darmstadt
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jeffrey S. A. Stringer
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Brice Gaudilliere
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P. Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Martin S. Angst
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Anisur Rahman
- Maternal and Child Health Division, International Centre for Diarrhoeal Disease Research, Dhaka, Bangladesh
| | - Abdullah H. Baqui
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fyezah Jehan
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Muhammad Imran Nisar
- Department of Pediatrics and Child Health, Faculty of Health Sciences, Medical College, The Aga Khan University, Karachi, Pakistan
| | - Bellington Vwalika
- Department of Obstetrics and Gynecology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Obstetrics and Gynaecology, University of Zambia School of Medicine, Lusaka, Zambia
| | - Sunil Sazawal
- Centre for Public Health Kinetics, New Delhi, Delhi, India
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Gary M. Shaw
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - David K. Stevenson
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
11
|
Hong J, Kumar S. Circulating biomarkers associated with placental dysfunction and their utility for predicting fetal growth restriction. Clin Sci (Lond) 2023; 137:579-595. [PMID: 37075762 PMCID: PMC10116344 DOI: 10.1042/cs20220300] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
Fetal growth restriction (FGR) leading to low birth weight (LBW) is a major cause of neonatal morbidity and mortality worldwide. Normal placental development involves a series of highly regulated processes involving a multitude of hormones, transcription factors, and cell lineages. Failure to achieve this leads to placental dysfunction and related placental diseases such as pre-clampsia and FGR. Early recognition of at-risk pregnancies is important because careful maternal and fetal surveillance can potentially prevent adverse maternal and perinatal outcomes by judicious pregnancy surveillance and careful timing of birth. Given the association between a variety of circulating maternal biomarkers, adverse pregnancy, and perinatal outcomes, screening tests based on these biomarkers, incorporating maternal characteristics, fetal biophysical or circulatory variables have been developed. However, their clinical utility has yet to be proven. Of the current biomarkers, placental growth factor and soluble fms-like tyrosine kinase 1 appear to have the most promise for placental dysfunction and predictive utility for FGR.
Collapse
Affiliation(s)
- Jesrine Hong
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland 4101, Australia
- Department of Obstetrics and Gynecology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- School of Medicine, The University of Queensland, Herston, Queensland 4006, Australia
| | - Sailesh Kumar
- Mater Research Institute, University of Queensland, Level 3, Aubigny Place, Raymond Terrace, South Brisbane, Queensland 4101, Australia
- School of Medicine, The University of Queensland, Herston, Queensland 4006, Australia
| |
Collapse
|
12
|
Elhakeem A, Ronkainen J, Mansell T, Lange K, Mikkola TM, Mishra BH, Wahab RJ, Cadman T, Yang T, Burgner D, Eriksson JG, Järvelin MR, Gaillard R, Jaddoe VWV, Lehtimäki T, Raitakari OT, Saffery R, Wake M, Wright J, Sebert S, Lawlor DA. Effect of common pregnancy and perinatal complications on offspring metabolic traits across the life course: a multi-cohort study. BMC Med 2023; 21:23. [PMID: 36653824 PMCID: PMC9850719 DOI: 10.1186/s12916-022-02711-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 12/14/2022] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Common pregnancy and perinatal complications are associated with offspring cardiometabolic risk factors. These complications may influence multiple metabolic traits in the offspring and these associations might differ with offspring age. METHODS We used data from eight population-based cohort studies to examine and compare associations of pre-eclampsia (PE), gestational hypertension (GH), gestational diabetes (GD), preterm birth (PTB), small (SGA) and large (LGA) for gestational age (vs. appropriate size for gestational age (AGA)) with up to 167 plasma/serum-based nuclear magnetic resonance-derived metabolic traits encompassing lipids, lipoproteins, fatty acids, amino acids, ketones, glycerides/phospholipids, glycolysis, fluid balance, and inflammation. Confounder-adjusted regression models were used to examine associations (adjusted for maternal education, parity age at pregnancy, ethnicity, pre/early pregnancy body mass index and smoking, and offspring sex and age at metabolic trait assessment), and results were combined using meta-analysis by five age categories representing different periods of the offspring life course: neonates (cord blood), infancy (mean ages: 1.1-1.6 years), childhood (4.2-7.5 years); adolescence (12.0-16.0 years), and adulthood (22.0-67.8 years). RESULTS Offspring numbers for each age category/analysis varied from 8925 adults (441 PTB) to 1181 infants (135 GD); 48.4% to 60.0% were females. Pregnancy complications (PE, GH, GD) were each associated with up to three metabolic traits in neonates (P≤0.001) with some evidence of persistence to older ages. PTB and SGA were associated with 32 and 12 metabolic traits in neonates respectively, which included an adjusted standardised mean difference of -0.89 standard deviation (SD) units for albumin with PTB (95% CI: -1.10 to -0.69, P=1.3×10-17) and -0.41 SD for total lipids in medium HDL with SGA (95% CI: -0.56 to -0.25, P=2.6×10-7), with some evidence of persistence to older ages. LGA was inversely associated with 19 metabolic traits including lower levels of cholesterol, lipoproteins, fatty acids, and amino acids, with associations emerging in adolescence, (e.g. -0.11 SD total fatty acids, 95% CI: -0.18 to -0.05, P=0.0009), and attenuating with older age across adulthood. CONCLUSIONS These reassuring findings suggest little evidence of wide-spread and long-term impact of common pregnancy and perinatal complications on offspring metabolic traits, with most associations only observed for newborns rather than older ages, and for perinatal rather than pregnancy complications.
Collapse
Affiliation(s)
- Ahmed Elhakeem
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK.
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK.
| | - Justiina Ronkainen
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Toby Mansell
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Katherine Lange
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Tuija M Mikkola
- Folkhälsan Research Center, Helsinki, Finland
- Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Binisha H Mishra
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Rama J Wahab
- Department of Paediatrics, Erasmus MC-University Medical Centre Rotterdam, Rotterdam, Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Centre, Rotterdam, Netherlands
| | - Tim Cadman
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Section of Epidemiology, Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Tiffany Yang
- Bradford Institute for Health Research, Bradford Teaching Hospitals National Health Service Foundation Trust, Bradford, UK
| | - David Burgner
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Department of Paediatrics, Monash University, Clayton, VIC, Australia
| | - Johan G Eriksson
- Folkhälsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Obstetrics & Gynecology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Singapore Institute for Clinical Sciences (SICS), Agency for Science and Technology (A*STAR), Singapore, Singapore
| | - Marjo-Riitta Järvelin
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Romy Gaillard
- Department of Paediatrics, Erasmus MC-University Medical Centre Rotterdam, Rotterdam, Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Centre, Rotterdam, Netherlands
| | - Vincent W V Jaddoe
- Department of Paediatrics, Erasmus MC-University Medical Centre Rotterdam, Rotterdam, Netherlands
- The Generation R Study Group, Erasmus MC, University Medical Centre, Rotterdam, Netherlands
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere, Finland
| | - Olli T Raitakari
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - Richard Saffery
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
| | - Melissa Wake
- Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia
- Liggins Institute, University of Auckland, Auckland, New Zealand
| | - John Wright
- Bradford Institute for Health Research, Bradford Teaching Hospitals National Health Service Foundation Trust, Bradford, UK
| | - Sylvain Sebert
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit at the University of Bristol, Bristol, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
- NIHR Bristol Biomedical Research Centre, Bristol, UK
| |
Collapse
|
13
|
Zhong Z, Yang Q, Li C, Chen X, Zhou F. A global perspective of correlation between maternal blood lead levels and risks of preeclampsia: An updated systematic review and meta-analysis. Front Public Health 2022; 10:1072052. [PMID: 36620238 PMCID: PMC9816335 DOI: 10.3389/fpubh.2022.1072052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Background Preeclampsia (PE) is a specific hypertensive disorder in pregnancy. Lead (Pb) is a heavy metal that affects women's reproductive health. However, it is unclear whether lead exposure during can predispose maternal risk of developing preeclampsia. This systematic review and meta-analysis study aimed to explore the association. Methods We searched studies from three databases (PubMed, Web of Science, Embase). Only case-control, cross-sectional, and cohort studies reporting maternal blood lead levels (BLL) and PE were included from database inception to 31st July 2022. Pregnant women with blood lead levels measured were eligible. Those healthy pregnant women who did not develop preeclampsia were assessed as comparators. Letters, comments, case reports, and reviews were excluded. Newcastle-Ottawa Scale (NOS) and its adaptive form were applied for assessment. The random-effects method (REM) was applied to calculate the standardized mean difference (SMD) with a 95% confidence interval (CI). Stata 16.0 and RevMan 5.3 were the software used for data extraction and analysis. Results 25 studies out of 1,808 articles made the finalist for systematic reviews, of which 21 underwent further quantity analysis. A total of 1,533 preeclamptic women and 10,998 healthy pregnant controls were included in the meta-analysis. The overall result revealed that maternal lead exposure was significantly higher in women with preeclampsia (SMD: 1.06, 95% CI 0.69, 1.43); (I 2 = 96.40%; P = 0.000). Conclusion This study demonstrates that maternal lead exposure is associated with preeclampsia during pregnancy. The association is present even in low blood lead levels. The conclusion should be taken seriously and women should avoid unexpected exposure to a lead-containing environment as much as possible. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=347220, identifier: CRD42022347220.
Collapse
Affiliation(s)
- Zixing Zhong
- Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China
| | - Qingmei Yang
- Graduate School, Bengbu Medical University, Bengbu, Anhui, China,Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, China
| | - Chu Li
- The Second Clinical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaohong Chen
- Department of Pediatrics, Central Hospital of Haining, Haining, Zhejiang, China,*Correspondence: Xiaohong Chen ✉
| | - Feifei Zhou
- Center for Reproductive Medicine, Department of Traditional Chinese Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, China,Feifei Zhou ✉
| |
Collapse
|
14
|
Cindrova-Davies T, Sferruzzi-Perri AN. Human placental development and function. Semin Cell Dev Biol 2022; 131:66-77. [PMID: 35393235 DOI: 10.1016/j.semcdb.2022.03.039] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/30/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022]
Abstract
The placenta is a transient fetal organ that plays a critical role in the health and wellbeing of both the fetus and its mother. Functionally, the placenta sustains the growth of the fetus as it facilitates delivery of oxygen and nutrients and removal of waste products. Not surprisingly, defective early placental development is the primary cause of common disorders of pregnancy, including recurrent miscarriage, fetal growth restriction, pre-eclampsia and stillbirth. Adverse pregnancy conditions will also affect the life-long health of the fetus via developmental programming[1]. Despite its critical importance in reproductive success and life-long health, our understanding of placental development is not extensive, largely due to ethical limitations to studying early or chronological placental development, lack of long-term in vitro models, or comparative animal models. In this review, we examine current knowledge of early human placental development, discuss the critical role of the maternal endometrium and of the fetal-maternal dialogue in pregnancy success, and we explore the latest models of trophoblast and endometrial stem cells. In addition, we discuss the role of oxygen in placental formation and function, how nutrient delivery is mediated during the periods of histotrophic nutrition (uptake of uterine secretions) and haemotrophic nutrition (exchange between the maternal and fetal circulations), and how placental endocrine function facilitates fetal growth and development.
Collapse
Affiliation(s)
- Tereza Cindrova-Davies
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
15
|
de Alwis N, Beard S, Binder NK, Pritchard N, Kaitu’u-Lino TJ, Walker SP, Stock O, Groom K, Petersen S, Henry A, Said JM, Seeho S, Kane SC, Tong S, Hui L, Hannan NJ. Placental OLAH Levels Are Altered in Fetal Growth Restriction, Preeclampsia and Models of Placental Dysfunction. Antioxidants (Basel) 2022; 11:1677. [PMID: 36139751 PMCID: PMC9495588 DOI: 10.3390/antiox11091677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Previously, we identified elevated transcripts for the gene Oleoyl-ACP Hydrolase (OLAH) in the maternal circulation of pregnancies complicated by preterm fetal growth restriction. As placental dysfunction is central to the pathogenesis of both fetal growth restriction and preeclampsia, we aimed to investigate OLAH levels and function in the human placenta. We assessed OLAH mRNA expression (qPCR) throughout pregnancy, finding placental expression increased as gestation progressed. OLAH mRNA and protein levels (Western blot) were elevated in placental tissue from cases of preterm preeclampsia, while OLAH protein levels in placenta from growth-restricted pregnancies were comparatively reduced in the preeclamptic cohort. OLAH expression was also elevated in placental explant tissue, but not isolated primary cytotrophoblast cultured under hypoxic conditions (as models of placental dysfunction). Further, we discovered that silencing cytotrophoblast OLAH reduced the expression of pro- and anti-apoptosis genes, BAX and BCL2, placental growth gene, IGF2, and oxidative stress gene, NOX4. Collectively, these findings suggest OLAH could play a role in placental dysfunction and may be a therapeutic target for mitigating diseases associated with this vital organ. Further research is required to establish the role of OLAH in the placenta, and whether these changes may be a maternal adaptation or consequence of disease.
Collapse
Affiliation(s)
- Natasha de Alwis
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Northern Health, Epping, VIC 3076, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sally Beard
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Northern Health, Epping, VIC 3076, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Natalie K. Binder
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Natasha Pritchard
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Tu’uhevaha J. Kaitu’u-Lino
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Susan P. Walker
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Owen Stock
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Katie Groom
- Liggins Institute, University of Auckland, Auckland 1023, New Zealand
| | - Scott Petersen
- Centre for Maternal Fetal Medicine, Mater Mothers’ Hospital, South Brisbane, QLD 4101, Australia
| | - Amanda Henry
- Discipline of Women’s Health, School of Clinical Medicine, UNSW Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Joanne M. Said
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Maternal Fetal Medicine, Joan Kirner Women’s & Children’s Sunshine Hospital, St Albans, VIC 3021, Australia
| | - Sean Seeho
- Women and Babies Research, Sydney Medical School-Northern, Faculty of Medicine and Health, University of Sydney, St Leonards, NSW 2065, Australia
| | - Stefan C. Kane
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Maternal Fetal Medicine, Royal Women’s Hospital, Parkville, VIC 3052, Australia
| | - Stephen Tong
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
- Translational Obstetrics Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Lisa Hui
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Northern Health, Epping, VIC 3076, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Natalie J. Hannan
- Therapeutics Discovery and Vascular Function in Pregnancy Group, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
- Northern Health, Epping, VIC 3076, Australia
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
16
|
Rau A, Passet B, Castille J, Daniel-Carlier N, Asset A, Lecardonnel J, Moroldo M, Jaffrézic F, Laloë D, Moazami-Goudarzi K, Vilotte JL. Potential genetic robustness of Prnp and Sprn double knockout mouse embryos towards ShRNA-lentiviral inoculation. Vet Res 2022; 53:54. [PMID: 35799279 PMCID: PMC9264527 DOI: 10.1186/s13567-022-01075-4] [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: 05/06/2022] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
The Shadoo and PrP prion protein family members are thought to be functionally related, but previous knockdown/knockout experiments in early mouse embryogenesis have provided seemingly contradictory results. In particular, Shadoo was found to be indispensable in the absence of PrP in knockdown analyses, but a double-knockout of the two had little phenotypic impact. We investigated this apparent discrepancy by comparing transcriptomes of WT, Prnp0/0 and Prnp0/0Sprn0/0 E6.5 mouse embryos following inoculation by Sprn- or Prnp-ShRNA lentiviral vectors. Our results suggest the possibility of genetic adaptation in Prnp0/0Sprn0/0 mice, thus providing a potential explanation for their previously observed resilience.
Collapse
Affiliation(s)
- Andrea Rau
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France. .,BioEcoAgro Joint Research Unit, INRAE, Université de Liège, Université de Lille, Université de Picardie Jules Verne, 80203, Péronne, France.
| | - Bruno Passet
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Johan Castille
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | | | - Alexandre Asset
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Jérome Lecardonnel
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Marco Moroldo
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Florence Jaffrézic
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Denis Laloë
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | | | - Jean-Luc Vilotte
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| |
Collapse
|
17
|
de Alwis N, Beard S, Binder NK, Pritchard N, Tong S, Kaitu'u-Lino TJ, Hannan NJ. Placental DAAM2 is unaltered in preeclampsia, but upregulated by treatment with proton pump inhibitors. Pregnancy Hypertens 2022; 30:13-20. [DOI: 10.1016/j.preghy.2022.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 11/24/2022]
|
18
|
Tong S, Walker SP, Keenan E, MacDonald TM, Taylor R, McCowan LME, Kaitu'u-Lino TJ. Circulating serine peptidase inhibitor Kunitz type 1 (SPINT1) in the second trimester is reduced among pregnancies that end in low birthweight neonates: cohort study of 2006 pregnancies. Am J Obstet Gynecol MFM 2022; 4:100618. [PMID: 35331972 DOI: 10.1016/j.ajogmf.2022.100618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 10/18/2022]
Affiliation(s)
- Stephen Tong
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, 163 Studley Rd., Heidelberg 3084, Melbourne, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
| | - Susan P Walker
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, 163 Studley Rd., Heidelberg 3084, Melbourne, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Emerson Keenan
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, 163 Studley Rd., Heidelberg 3084, Melbourne, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Teresa M MacDonald
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, 163 Studley Rd., Heidelberg 3084, Melbourne, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Rennae Taylor
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Lesley M E McCowan
- Department of Obstetrics and Gynaecology, University of Auckland, Auckland, New Zealand
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Australia; Mercy Perinatal, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| |
Collapse
|
19
|
Evidence-Based Screening, Diagnosis and Management of Fetal Growth Restriction: Challenges and Confusions. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
20
|
The Update of Fetal Growth Restriction Associated with Biomarkers. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
21
|
Circulating SPINT1 Is Reduced in a Preeclamptic Cohort with Co-Existing Fetal Growth Restriction. J Clin Med 2022; 11:jcm11040901. [PMID: 35207174 PMCID: PMC8877863 DOI: 10.3390/jcm11040901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/01/2022] [Accepted: 02/06/2022] [Indexed: 02/01/2023] Open
Abstract
Fetal growth restriction (FGR), when undetected antenatally, is the biggest risk factor for preventable stillbirth. Maternal circulating SPINT1 is reduced in pregnancies, which ultimately deliver small for gestational age (SGA) infants at term (birthweight < 10th centile), compared to appropriate for gestational age (AGA) infants (birthweight ≥ 10th centile). SPINT1 is also reduced in FGR diagnosed before 34 weeks’ gestation. We hypothesised that circulating SPINT1 would be decreased in co-existing preterm preeclampsia and FGR. Plasma SPINT1 was measured in samples obtained from two double-blind, randomised therapeutic trials. In the Preeclampsia Intervention with Esomeprazole trial, circulating SPINT1 was decreased in women with preeclampsia who delivered SGA infants (n = 75, median = 18,857 pg/mL, IQR 10,782–29,890 pg/mL, p < 0.0001), relative to those delivering AGA (n = 22, median = 40,168 pg/mL, IQR 22,342–75,172 pg/mL). This was confirmed in the Preeclampsia Intervention 2 with metformin trial where levels of SPINT1 in maternal circulation were reduced in SGA pregnancies (n = 95, median = 57,764 pg/mL, IQR 42,212–91,356 pg/mL, p < 0.0001) compared to AGA controls (n = 40, median = 107,062 pg/mL, IQR 70,183–176,532 pg/mL). Placental Growth Factor (PlGF) and sFlt-1 were also measured. PlGF was significantly reduced in the SGA pregnancies, while ratios of sFlt-1/SPINT1 and sFlt1/PlGF were significantly increased. This is the first study to demonstrate significantly reduced SPINT1 in co-existing FGR and preeclamptic pregnancies.
Collapse
|
22
|
Andres F, Wong GP, Walker SP, MacDonald TM, Keenan E, Cannon P, Nguyen TV, Hannan NJ, Tong S, Kaitu'u-Lino TJ. A disintegrin and metalloproteinase 12 (ADAM12) is reduced at 36 weeks' gestation in pregnancies destined to deliver small for gestational age infants. Placenta 2021; 117:1-4. [PMID: 34768162 DOI: 10.1016/j.placenta.2021.11.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/18/2021] [Accepted: 11/01/2021] [Indexed: 11/26/2022]
Abstract
First trimester circulating ADAM12 is reduced in fetal growth restriction (FGR) and preeclampsia. We measured plasma ADAM12 at 36 weeks' gestation preceding diagnosis of term preeclampsia or delivery of a small for gestational age (SGA; birthweight <10th centile) infant in two independent cohorts (Cohort 1 90 SGA, 41 preeclampsia, 862 controls; Cohort 2121 SGA 23 preeclampsia; 190 controls). ADAM12 was reduced with SGA in both cohorts (p = 0.0015 and 0.011 respectively), and further reduced with birthweight <5th centile (p = 0.0013 and 0.0058 respectively). This validates ADAM12 as an SGA biomarker near term. Circulating ADAM12 preceding preeclampsia was not consistently altered.
Collapse
Affiliation(s)
- Faith Andres
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Georgia P Wong
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Susan P Walker
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Teresa M MacDonald
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Emerson Keenan
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Tuong-Vi Nguyen
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Natalie J Hannan
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Stephen Tong
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia.
| |
Collapse
|
23
|
NR4A2 expression is not altered in placentas from cases of growth restriction or preeclampsia, but is reduced in hypoxic cytotrophoblast. Sci Rep 2021; 11:20670. [PMID: 34667209 PMCID: PMC8526588 DOI: 10.1038/s41598-021-00192-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/29/2021] [Indexed: 11/21/2022] Open
Abstract
Nuclear Receptor Subfamily 4 Group A Member 2 (NR4A2) transcripts are elevated in the circulation of individuals whose pregnancies are complicated by preterm fetal growth restriction (FGR). In this paper, we show that the cases with preeclampsia (PE) have increased circulating NR4A2 transcripts compared to those with normotensive FGR. We aimed to establish whether the dysfunctional placenta mirrors the increase in NR4A2 transcripts and further, to uncover the function of placental NR4A2. NR4A2 expression was detected in preterm and term placental tissue; expressed higher at term. NR4A2 mRNA expression and protein were not altered in placentas from preterm FGR or PE pregnancies. Hypoxia (1% O2 compared to 8% O2) significantly reduced cytotrophoblast NR4A2 mRNA expression, but not placental explant NR4A2 expression. Silencing cytotrophoblast NR4A2 expression under hypoxia (via short interfering (si)RNAs) did not alter angiogenic Placental Growth Factor, nor anti-angiogenic sFlt-1 mRNA expression or protein secretion, but increased expression of cellular antioxidant, oxidative stress, inflammatory, and growth genes. NR4A2 expression was also not altered in a model of tumour necrosis factor-α-induced endothelial dysfunction, or with pravastatin treatment. Further studies are required to identify the origin of the circulating transcripts in pathological pregnancies, and investigate the function of placental NR4A2.
Collapse
|
24
|
Circulating syndecan-1 is reduced in pregnancies with poor fetal growth and its secretion regulated by matrix metalloproteinases and the mitochondria. Sci Rep 2021; 11:16595. [PMID: 34400721 PMCID: PMC8367987 DOI: 10.1038/s41598-021-96077-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 07/30/2021] [Indexed: 11/08/2022] Open
Abstract
Fetal growth restriction is a leading cause of stillbirth that often remains undetected during pregnancy. Identifying novel biomarkers may improve detection of pregnancies at risk. This study aimed to assess syndecan-1 as a biomarker for small for gestational age (SGA) or fetal growth restricted (FGR) pregnancies and determine its molecular regulation. Circulating maternal syndecan-1 was measured in several cohorts; a large prospective cohort collected around 36 weeks’ gestation (n = 1206), a case control study from the Manchester Antenatal Vascular service (285 women sampled at 24–34 weeks’ gestation); two prospective cohorts collected on the day of delivery (36 + 3–41 + 3 weeks’ gestation, n = 562 and n = 405 respectively) and a cohort who delivered for preterm FGR (< 34 weeks). Circulating syndecan-1 was consistently reduced in women destined to deliver growth restricted infants and those delivering for preterm disease. Syndecan-1 secretion was reduced by hypoxia, and its loss impaired proliferation. Matrix metalloproteinases and mitochondrial electron transport chain inhibitors significantly reduced syndecan-1 secretion, an effect that was rescued by coadministration of succinate, a mitochondrial electron transport chain activator. In conclusion, circulating syndecan-1 is reduced among cases of term and preterm growth restriction and has potential for inclusion in multi-marker algorithms to improve detection of poorly grown fetuses.
Collapse
|
25
|
Selvaratnam RJ, Wallace EM, Wolfe R, Anderson PJ, Davey MA. Association Between Iatrogenic Delivery for Suspected Fetal Growth Restriction and Childhood School Outcomes. JAMA 2021; 326:145-153. [PMID: 34255007 PMCID: PMC8278267 DOI: 10.1001/jama.2021.8608] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
IMPORTANCE Timely delivery of infants suspected of having fetal growth restriction (FGR) is a balance between preventing stillbirth and minimizing prematurity, particularly because many infants with suspected FGR have normal growth. OBJECTIVE To explore the association between iatrogenic delivery for suspected FGR and childhood school outcomes. DESIGN, SETTING, AND PARTICIPANTS A retrospective whole-population cohort study linking perinatal data from births 32 weeks' or more gestation between January 1, 2003, to December 31, 2013, to developmental and educational test scores at preparatory school, and at school grades 3, 5, and 7 in Victoria, Australia. Follow-up was concluded in 2019. EXPOSURES Suspicion or nonsuspicion of FGR, presence or absence of iatrogenic delivery (defined as early induction of labor or cesarean delivery prior to labor) for suspected FGR, and presence or absence of small for gestational age (SGA). MAIN OUTCOMES AND MEASURES The coprimary outcomes were being in the bottom 10th percentile on 2 or more of 5 developmental domains at school entry and being below the national minimum standard on 2 or more of 5 educational domains in grades 3, 5, or 7. RESULTS In the birth population of 705 937 infants, the mean gestation at birth was 39.1 (SD, 1.5) weeks and the mean birth weight was 3426 (SD, 517) grams. The birth population linked to 181 902 children with developmental results and 425 717 children with educational results. Compared with infants with severe SGA (birth weight <3rd percentile) not suspected of having FGR, infants with severe SGA delivered for suspected FGR were born earlier (mean gestation, 37.9 weeks vs 39.4 weeks). They also had a significantly increased risk of poor developmental outcome at school entry (16.2% vs 12.7%; absolute difference, 3.5% [95% CI, 0.5%-6.5%]); adjusted odds ratio [aOR], 1.36 [95% CI, 1.07-1.74]) and poor educational outcomes in grades 3, 5, and 7 (for example, in grade 7: 13.4% vs 10.5%; absolute difference, 2.9% [95% CI, 0.4%-5.5%]); aOR, 1.33 [95% CI, 1.04-1.70]). There was no significant difference between infants with normal growth (birth weight ≥10th percentile) delivered for suspected FGR and those not suspected of having FGR in developmental outcome (8.6% vs 8.1%; absolute difference, 0.5% [95% CI, -1.1% to 2.0%]); aOR, 1.17 [95% CI, 0.95-1.45]) or educational outcome in grade 3, 5 or 7, despite being born earlier (mean gestation, 38.0 weeks vs 39.1 weeks). CONCLUSIONS AND RELEVANCE In this exploratory study conducted in Victoria, Australia, iatrogenic delivery of infants with severe SGA due to suspected FGR was associated with poorer school outcomes compared with infants with severe SGA not suspected of having FGR. Iatrogenic delivery of infants with normal growth due to suspected FGR was not associated with poorer school outcomes compared with infants with normal growth not suspected of having FGR.
Collapse
Affiliation(s)
- Roshan John Selvaratnam
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
- Safer Care Victoria, Department of Health, Victorian Government, Victoria, Australia
| | - Euan Morrison Wallace
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
- Safer Care Victoria, Department of Health, Victorian Government, Victoria, Australia
| | - Rory Wolfe
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Peter John Anderson
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, Victoria, Australia
- School of Psychological Sciences, Monash University, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Mary-Ann Davey
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
- Safer Care Victoria, Department of Health, Victorian Government, Victoria, Australia
| |
Collapse
|
26
|
Murphy CN, Walker SP, MacDonald TM, Keenan E, Hannan NJ, Wlodek ME, Myers J, Briffa JF, Romano T, Roddy Mitchell A, Whigham CA, Cannon P, Nguyen TV, Kandel M, Pritchard N, Tong S, Kaitu’u-Lino TJ. Elevated Circulating and Placental SPINT2 Is Associated with Placental Dysfunction. Int J Mol Sci 2021; 22:7467. [PMID: 34299087 PMCID: PMC8305184 DOI: 10.3390/ijms22147467] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/08/2021] [Accepted: 07/08/2021] [Indexed: 12/04/2022] Open
Abstract
Biomarkers for placental dysfunction are currently lacking. We recently identified SPINT1 as a novel biomarker; SPINT2 is a functionally related placental protease inhibitor. This study aimed to characterise SPINT2 expression in placental insufficiency. Circulating SPINT2 was assessed in three prospective cohorts, collected at the following: (1) term delivery (n = 227), (2) 36 weeks (n = 364), and (3) 24-34 weeks' (n = 294) gestation. SPINT2 was also measured in the plasma and placentas of women with established placental disease at preterm (<34 weeks) delivery. Using first-trimester human trophoblast stem cells, SPINT2 expression was assessed in hypoxia/normoxia (1% vs. 8% O2), and following inflammatory cytokine treatment (TNFα, IL-6). Placental SPINT2 mRNA was measured in a rat model of late-gestational foetal growth restriction. At 36 weeks, circulating SPINT2 was elevated in patients who later developed preeclampsia (p = 0.028; median = 2233 pg/mL vs. controls, median = 1644 pg/mL), or delivered a small-for-gestational-age infant (p = 0.002; median = 2109 pg/mL vs. controls, median = 1614 pg/mL). SPINT2 was elevated in the placentas of patients who required delivery for preterm preeclampsia (p = 0.025). Though inflammatory cytokines had no effect, hypoxia increased SPINT2 in cytotrophoblast stem cells, and its expression was elevated in the placental labyrinth of growth-restricted rats. These findings suggest elevated SPINT2 is associated with placental insufficiency.
Collapse
Affiliation(s)
- Ciara N. Murphy
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Susan P. Walker
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Teresa M. MacDonald
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Emerson Keenan
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Natalie J. Hannan
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Mary E. Wlodek
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- The Department of Anatomy and Physiology, The University of Melbourne, VIC 3010, Australia;
| | - Jenny Myers
- Manchester Academic Health Science Centre, St Mary’s Hospital, University of Manchester, Manchester M13 OJH, UK;
| | - Jessica F. Briffa
- The Department of Anatomy and Physiology, The University of Melbourne, VIC 3010, Australia;
| | - Tania Romano
- The Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia;
| | - Alexandra Roddy Mitchell
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Carole-Anne Whigham
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Ping Cannon
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Tuong-Vi Nguyen
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Manju Kandel
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Natasha Pritchard
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Stephen Tong
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| | - Tu’uhevaha J. Kaitu’u-Lino
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, The University of Melbourne, Heidelberg, VIC 3084, Australia; (S.P.W.); (T.M.M.); (E.K.); (N.J.H.); (M.E.W.); (A.R.M.); (C.-A.W.); (P.C.); (T.-V.N.); (M.K.); (N.P.); (S.T.); (T.J.K.-L.)
- Mercy Perinatal, Mercy Hospital for Women, Heidelberg, VIC 3084, Australia
| |
Collapse
|
27
|
Kaitu'u-Lino TJ, Tong S, Walker SP, MacDonald TM, Cannon P, Nguyen TV, Sadananthan SA, Tint MT, Ong YY, Ling LS, Gluckman PD, Chong YS, Godfrey KM, Chan SY, Tan KH, Lee YS, Michael N, Eriksson JG, Wlodek ME. Maternal circulating SPINT1 is reduced in small-for-gestational age pregnancies at 26 weeks: Growing up in Singapore towards health outcomes (GUSTO) cohort study. Placenta 2021; 110:24-28. [PMID: 34102451 DOI: 10.1016/j.placenta.2021.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/11/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
Fetal growth restriction arising from placental insufficiency is a leading cause of stillbirth. We recently identified low maternal circulating SPINT1 concentrations as a novel biomarker of poor fetal growth. Here we measured SPINT1 in a prospective cohort in Singapore. Circulating SPINT1 concentrations were significantly lower among 141 pregnant women destined to deliver small-for-gestational age infants (birthweight <10th centile), compared to 772 controls (p < 0.01) at as early as 26 weeks' gestation. There were no correlations between infant body composition and circulating SPINT1 concentrations at 26 weeks. This provides validation that low maternal SPINT1 concentration is associated with poor fetal growth.
Collapse
Affiliation(s)
- Tu'uhevaha J Kaitu'u-Lino
- Translational Obstetrics Group, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia.
| | - Stephen Tong
- Translational Obstetrics Group, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Susan P Walker
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Teresa M MacDonald
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Victoria, Australia; Mercy Perinatal, Mercy Hospital for Women, Victoria, Australia
| | - Ping Cannon
- Translational Obstetrics Group, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Victoria, Australia
| | - Tuong-Vi Nguyen
- Translational Obstetrics Group, Mercy Hospital for Women, 163 Studley Road, Heidelberg, 3084, Victoria, Australia; The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Victoria, Australia
| | - Suresh Anand Sadananthan
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Mya-Thway Tint
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yi Ying Ong
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Loy See Ling
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore, 229899; Duke-NUS Medical School, Singapore, 169857
| | - Peter D Gluckman
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore; Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Yap-Seng Chong
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Unit and NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, UK
| | - Shiao-Yng Chan
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kok Hian Tan
- Department of Maternal FetaL Medicine KK Women's and Children's Hospital Singapore, Singapore
| | - Yung Seng Lee
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Navin Michael
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Johan G Eriksson
- Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Folkhälsan Research Center, Helsinki, Finland
| | - Mary E Wlodek
- The Department of Obstetrics and Gynaecology, Mercy Hospital for Women, University of Melbourne, Victoria, Australia; Singapore Institute of Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore; Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
28
|
Napso T, Zhao X, Lligoña MI, Sandovici I, Kay RG, George AL, Gribble FM, Reimann F, Meek CL, Hamilton RS, Sferruzzi-Perri AN. Placental secretome characterization identifies candidates for pregnancy complications. Commun Biol 2021; 4:701. [PMID: 34103657 PMCID: PMC8187406 DOI: 10.1038/s42003-021-02214-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 05/12/2021] [Indexed: 11/09/2022] Open
Abstract
Alterations in maternal physiological adaptation during pregnancy lead to complications, including abnormal birthweight and gestational diabetes. Maternal adaptations are driven by placental hormones, although the full identity of these is lacking. This study unbiasedly characterized the secretory output of mouse placental endocrine cells and examined whether these data could identify placental hormones important for determining pregnancy outcome in humans. Secretome and cell peptidome analyses were performed on cultured primary trophoblast and fluorescence-activated sorted endocrine trophoblasts from mice and a placental secretome map was generated. Proteins secreted from the placenta were detectable in the circulation of mice and showed a higher relative abundance in pregnancy. Bioinformatic analyses showed that placental secretome proteins are involved in metabolic, immune and growth modulation, are largely expressed by human placenta and several are dysregulated in pregnancy complications. Moreover, proof-of-concept studies found that secreted placental proteins (sFLT1/MIF and ANGPT2/MIF ratios) were increased in women prior to diagnosis of gestational diabetes. Thus, placental secretome analysis could lead to the identification of new placental biomarkers of pregnancy complications.
Collapse
Affiliation(s)
- Tina Napso
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Xiaohui Zhao
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Marta Ibañez Lligoña
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Ionel Sandovici
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Department of Obstetrics and Gynaecology, The Rosie Hospital, Cambridge, UK
| | - Richard G Kay
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Amy L George
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona M Gribble
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Frank Reimann
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Claire L Meek
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Russell S Hamilton
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
29
|
Association of high maternal blood alpha-fetoprotein level with risk of delivering small for gestational age: a meta-analysis. Pediatr Res 2021; 89:1742-1750. [PMID: 32919396 DOI: 10.1038/s41390-020-01124-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/16/2020] [Accepted: 08/03/2020] [Indexed: 11/09/2022]
Abstract
BACKGROUND Early identification of pregnant women at risk of delivering small-for-gestational-age (SGA) infants is required to reduce the rates of mortality and morbidity in their whole life. This meta-analysis was performed to determine whether women with higher blood alpha-fetoprotein (AFP) levels are at increased risk of SGA. METHODS Studies identified by searching 11 databases, including PubMed, were assessed using the Newcastle-Ottawa Scale. Subgroup and meta-regression analyses and sensitivity analysis removing a potential outlier were performed. Publication bias was assessed using Egger's test. RESULTS A total of 39 good-quality cohort studies involving 93,968 women and their newborn infants or fetuses ensured both internal and external validity. Relative risk of SGA among women with higher in comparison to lower blood AFP levels was 2.021 (95% CI: 1.751-2.334). Maternal blood AFP levels showed a dose-response relationship with risk of SGA. Relative risk was higher with diagnosis of SGA by ultrasound than actual birth weight (P = 0.004). Sources of heterogeneity, subgroups, confounding factors, or potential outliers did not alter the interpretations without publication bias (P = 0.320). CONCLUSIONS Pregnant women with higher blood AFP levels are at increased risk of SGA compared to those with lower blood AFP levels. IMPACT Pregnant women with higher blood alpha-fetoprotein levels (AFP) levels are at increased risk of small for gestational age (SGA) compared to those with lower blood AFP levels. High-quality evidence showed a dose-response relationship of maternal blood AFP levels with risk of delivering SGA and was robust to sources of heterogeneity, subgroups, confounding factors, potential outliers, or publication bias. Politically and practically, monitoring of maternal blood AFP level is strongly recommended to identify women at risk of delivering SGA.
Collapse
|
30
|
Selvaratnam RJ, Wallace EM, Hunt RW, Davey MA. Preventing harm: A balance measure for improving the detection of fetal growth restriction. Aust N Z J Obstet Gynaecol 2021; 61:715-721. [PMID: 33772758 DOI: 10.1111/ajo.13340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 02/23/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Increasing the detection of fetal growth restriction (FGR), while reducing stillbirth, also leads to unnecessary early intervention, and associated morbidity, for normally grown babies who are incorrectly suspected of FGR. AIMS We sought to design a balance measure that addresses the specificity of FGR detection. METHODS A retrospective cohort study on all singleton births ≥32 weeks gestation in 2016 and 2017 in Victoria. We compared two balance measures for the detection of FGR, defined as the proportion of all babies iatrogenically delivered before 39 weeks gestation for suspected FGR that had a birthweight ≥10th centile (balance measure 1) or ≥25th centile (balance measure 2). Hospital level performance on each balance measure was derived and compared to an existing performance measure for severe FGR detection in Victoria. RESULTS Of the 38 hospitals analysed, 12 (32%) had a favourable performance on an existing indicator of FGR detection, seven (18%) hospitals had a favourable performance on balance measure 1, and 15 (39%) had a favourable performance on balance measure 2. There was a moderate correlation between hospital performance on the existing indicator and on balance measure 1 (r = 0.447, P = 0.005) but not balance measure 2 (r = -0.063, P = 0.71). There was no difference in perinatal mortality between high performing hospitals and low performing hospitals. CONCLUSION Introducing a balance measure into routine reporting may bring greater awareness to the unintended harm associated with increased detection of FGR.
Collapse
Affiliation(s)
- Roshan J Selvaratnam
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia.,Safer Care Victoria, Department of Health and Human Services, Victorian Government, Melbourne, Victoria, Australia
| | - Euan M Wallace
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia.,Safer Care Victoria, Department of Health and Human Services, Victorian Government, Melbourne, Victoria, Australia
| | - Rodney W Hunt
- Safer Care Victoria, Department of Health and Human Services, Victorian Government, Melbourne, Victoria, Australia.,Department of Paediatrics, Monash University, Melbourne, Victoria, Australia.,Neonatal Research, Clinical Sciences, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Monash Newborn, Monash Health, Melbourne, Victoria, Australia
| | - Mary-Ann Davey
- The Ritchie Centre, Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia.,Safer Care Victoria, Department of Health and Human Services, Victorian Government, Melbourne, Victoria, Australia
| |
Collapse
|
31
|
Chou FS, Newton K, Wang PS. Quantifying Fetal Reprogramming for Biomarker Development in the Era of High-Throughput Sequencing. Genes (Basel) 2021; 12:329. [PMID: 33668810 PMCID: PMC7996299 DOI: 10.3390/genes12030329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 11/16/2022] Open
Abstract
Gestational hypertensive disorders continue to threaten the well-being of pregnant women and their offspring. The only current definitive treatment for gestational hypertensive disorders is delivery of the fetus. The optimal timing of delivery remains controversial. Currently, the available clinical tools do not allow for assessment of fetal stress in its early stages. Placental insufficiency and fetal growth restriction secondary to gestational hypertensive disorders have been shown to have long-term impacts on offspring health even into their adulthood, becoming one of the major focuses of research in the field of developmental origins of health and disease. Fetal reprogramming was introduced to describe the long-lasting effects of the toxic intrauterine environment on the growing fetus. With the advent of high-throughput sequencing, there have been major advances in research attempting to quantify fetal reprogramming. Moreover, genes that are found to be differentially expressed as a result of fetal reprogramming show promise in the development of transcriptional biomarkers for clinical use in detecting fetal response to placental insufficiency. In this review, we will review key pathophysiology in the development of placental insufficiency, existing literature on high-throughput sequencing in the study of fetal reprogramming, and considerations regarding research design from our own experience.
Collapse
Affiliation(s)
- Fu-Sheng Chou
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA;
| | - Krystel Newton
- Division of Neonatology, Department of Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA;
| | - Pei-Shan Wang
- PXT Research & Data Analytics, LLC, Rancho Cucamonga, CA 91739, USA;
| |
Collapse
|
32
|
Soluble syndecan-1 and glycosaminoglycans in preeclamptic and normotensive pregnancies. Sci Rep 2021; 11:4387. [PMID: 33623064 PMCID: PMC7902809 DOI: 10.1038/s41598-021-82972-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/27/2021] [Indexed: 12/23/2022] Open
Abstract
Preeclampsia, an important cause of maternal and fetal morbidity and mortality, is associated with increased sFLT1 levels and with structural and functional damage to the glycocalyx contributing to endothelial dysfunction. We investigated glycocalyx components in relation to preeclampsia in human samples. While soluble syndecan-1 and heparan sulphate were similar in plasma of preeclamptic and normotensive pregnant women, dermatan sulphate was increased and keratan sulphate decreased in preeclamptic women. Dermatan sulphate was correlated with soluble syndecan-1, and inversely correlated with blood pressure and activated partial thromboplastin time. To determine if syndecan-1 was a prerequisite for the sFlt1 induced increase in blood pressure in mice we studied the effect of sFlt1 on blood pressure and vascular contractile responses in syndecan-1 deficient and wild type male mice. The classical sFlt1 induced rise in blood pressure was absent in syndecan-1 deficient mice indicating that syndecan-1 is a prerequisite for sFlt1 induced increase in blood pressure central to preeclampsia. The results show that an interplay between syndecan-1 and dermatan sulphate contributes to sFlt1 induced blood pressure elevation in pre-eclampsia.
Collapse
|
33
|
Circulating Tissue Factor Pathway Inhibitor (TFPI) is increased preceding preeclampsia diagnosis and in established preeclampsia. Placenta 2021; 105:32-40. [PMID: 33529886 DOI: 10.1016/j.placenta.2021.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/08/2021] [Accepted: 01/19/2021] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Tissue Factor Pathway Inhibitor (TFPI) is a part of the extrinsic coagulation pathway, and highly expressed in the placenta. We aimed to assess its potential as a preeclampsia biomarker. METHODS Maternal plasma was prospectively collected at 36 weeks' gestation. Circulating TFPI was measured in a nested case-control group (39 women who developed preeclampsia, 98 controls), before being measured in a larger independent cohort along with Placental Growth Factor (PlGF; 41 who developed preeclampsia, 954 controls). Circulating TFPI was then measured in women with underlying vascular disease, and also assessed in the plasma and placentas from women with preterm preeclampsia (delivered at <34 weeks). RESULTS Circulating TFPI was significantly increased in women destined to develop preeclampsia in the case-control study, a finding that validated in Cohort 2, with median TFPI in the preeclampsia group being 42.3 ng/ml (IQR 30-51 ng/ml) compared to 30 ng/ml (IQR 23.1-38.6 ng/ml) in controls (p < 0.0001). The area under the receiver operator characteristic curve (AUC) was 0.70. PlGF was significantly reduced in the preeclampsia group, and a ratio of TFPI/PlGF had an improved AUC of 0.78. In women with underlying vascular disease who were later diagnosed with early onset preeclampsia, circulating TFPI was significantly increased with a 0.29 (95% CI 0.13-0.44) increase in logTFPI (adjusted for gestation and hypertensive status). Circulating and placental TFPI were significantly increased in women with preterm preeclampsia. DISCUSSION Circulating TFPI is increased in women preceding diagnosis of preeclampsia (at 36 weeks) and in women with preterm disease. TFPI may beneficially contribute to a multi-marker blood test to predict preeclampsia.
Collapse
|
34
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
35
|
Stillbirth in Australia 1: The road to now: Two decades of stillbirth research and advocacy in Australia. Women Birth 2020; 33:506-513. [PMID: 33092699 DOI: 10.1016/j.wombi.2020.09.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 11/23/2022]
Abstract
Stillbirth is a major public health problem with an enormous mortality burden and psychosocial impact on parents, families and the wider community both globally and in Australia. In 2015, Australia's late gestation stillbirth rate was over 30% higher than that of the best-performing countries globally, highlighting the urgent need for action. We present an overview of the foundations which led to the establishment of Australia's NHMRC Centre of Research Excellence in Stillbirth (Stillbirth CRE) in 2017 and highlight key activities in the following areas: Opportunities to expand and improve collaborations between research teams; Supporting the conduct and development of innovative, high quality, collaborative research that incorporates a strong parent voice; Promoting effective translation of research into health policy and/or practice; and the Regional and global work of the Stillbirth CRE. We highlight the first-ever Senate Inquiry into Stillbirth in Australia in 2018. These events ultimately led to the development of a National Stillbirth Action and Implementation Plan for Australia with the aims of reducing stillbirth rates by 20% over the next five years, reducing the disparity in stillbirth rates between advantaged and disadvantaged communities, and improving care for all families who experience this loss.
Collapse
|
36
|
Ormesher L, Warrander L, Liu Y, Thomas S, Simcox L, Smith GCS, Myers JE, Johnstone ED. Risk stratification for early-onset fetal growth restriction in women with abnormal serum biomarkers: a retrospective cohort study. Sci Rep 2020; 10:22259. [PMID: 33335122 PMCID: PMC7746767 DOI: 10.1038/s41598-020-78631-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/28/2020] [Indexed: 11/08/2022] Open
Abstract
Abnormal maternal serum biomarkers (AMSB), identified through the aneuploidy screening programme, are frequent incidental findings in pregnancy. They are associated with fetal growth restriction (FGR), but previous studies have not examined whether this association is with early-onset (< 34 weeks) or late-onset (> 34 weeks) FGR; as a result there is no consensus on management. The aims of this study were to determine the prevalence and phenotype of FGR in women with AMSB and test the predictive value of placental sonographic screening to predict early-onset FGR. 1196 pregnant women with AMSB underwent a 21-24 week "placental screen" comprising fetal and placental size, and uterine artery Doppler. Multivariable regression was used to calculate a predictive model for early-onset FGR (birthweight centile < 3rd/< 10th with absent umbilical end-diastolic flow, < 34 weeks). FGR prevalence was high (10.3%), however early-onset FGR was uncommon (2.3%). Placental screening effectively identified early-onset (area under the curve (AUC) 0.93, 95% confidence interval (CI) 0.87-1.00), but not late-onset FGR (AUC 0.70, 95% CI 0.64-0.75). Internal validation demonstrated robust performance for detection/exclusion of early-onset FGR. In this cohort, utilisation of our proposed algorithm with targeted fetal growth and Doppler surveillance, compared with universal comprehensive surveillance would have avoided 1044 scans, potentiating significant cost-saving for maternity services.
Collapse
Affiliation(s)
- L Ormesher
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- St Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - L Warrander
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- St Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Y Liu
- Monash University, Scenic Boulevard & Wellington Road, Clayton, 3800, Australia
| | - S Thomas
- St Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - L Simcox
- St Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - G C S Smith
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
- NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | - J E Myers
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
- St Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK
| | - E D Johnstone
- Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
- St Mary's Hospital, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester, UK.
| |
Collapse
|
37
|
Real-time Assessment of the Development and Function of the Placenta Across Gestation to Support Therapeutics in Pregnancy. Clin Ther 2020; 43:279-286. [PMID: 33246660 DOI: 10.1016/j.clinthera.2020.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/02/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022]
Abstract
The placenta is vital to the health and development of the fetus, serving to deliver oxygen and nutrients, facilitate the removal of waste products, and provide a barrier to pathogens and other harmful substances present in the maternal circulation. When these processes fail to operate normally, they can lead to complications of pregnancy such as preeclampsia or fetal growth restriction. The development of novel therapeutics for the mother, fetus, or placenta requires a mechanistic understanding of the development and functions of the placenta. For the obstetric clinician, being able to monitor the placenta throughout the pregnancy and to measure the impact of any treatment modality on the mother and the developing fetus are essential for providing the best possible care. The Eunice Kennedy Shriver National Institute of Child Health and Human Development at the National Institutes of Health has been a longtime supporter of research on the placenta. In 2014, the Human Placenta Project was initiated to help to drive an understanding of the biology of the human placenta and to facilitate the development of novel tools and approaches to allow for safe, noninvasive, real-time assessment of the placenta across pregnancy. Those efforts, along with others from around the globe, are showing promise. Although not yet ready for clinical application, these advances are moving the field forward and are certain to have a tremendous impact on the development and assessment of therapeutics designed for treating conditions of pregnancy.
Collapse
|