1
|
Chai L, Li S, Yin B, Zhu X, Zhu B, Wu K. Prevalence, risk factors, and adverse perinatal outcomes in Chinese women with preeclampsia: a large retrospective cohort study. JOURNAL OF HEALTH, POPULATION, AND NUTRITION 2025; 44:32. [PMID: 39920879 PMCID: PMC11806619 DOI: 10.1186/s41043-025-00778-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/29/2025] [Indexed: 02/09/2025]
Abstract
BACKGROUND Preeclampsia (PE) is the primary cause of maternal and neonatal morbidity and mortality. However, comprehensive studies on the related risk factors with PE and its effects on adverse perinatal outcomes are limited. This study aimed to evaluate the prevalence, risk factors, and adverse perinatal outcomes in Chinese women with preeclampsia. METHODS We conducted a retrospective cohort study from January 1, 2018, to December 31, 2019, which enrolled 38,496 women without preeclampsia (non-PE) and 1130 women with PE. Univariate and multivariate logistic regression models were used to determine the risk factors and adverse perinatal outcomes of PE. RESULTS Multivariate logistic regression models showed that maternal age > 35 years, pp-BMI overweight/obesity, excessive gestational weight gain, multiparity, twin pregnancy, IVF, cesarean section history, times of abortion history ≥ 2, GDM, and ICP were significantly associated with the risk of PE (all P < 0.05). Women with PE in singleton pregnancies were associated with an increased risk of maternal outcomes of cesarean section, and preterm birth, and a higher risk of neonatal outcomes of stillbirth, low birth weight, fetal distress, neonatal asphyxia, and neonatal unit admission, which were also observed in women with PE in twin pregnancies, except for stillbirth and neonatal asphyxia. CONCLUSION This study identified the risk factors and associated adverse perinatal outcomes of PE, which providing comprehensive evidence for clinicians to manage women at risk of PE.
Collapse
Affiliation(s)
- Lin Chai
- Department of Clinical Laboratory, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuai Li
- Department of Clinical Laboratory, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Binbin Yin
- Department of Clinical Laboratory, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaojun Zhu
- Department of Obstetrics, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bo Zhu
- Department of Clinical Laboratory, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Kaiqi Wu
- Department of Clinical Laboratory, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Lokki AI, Triebwasser M, Daly E, Kurki MI, Perola M, Auro K, Salmon JE, Anuja J, Daly M, Atkinson JP, Laivuori H, Meri S. Understanding rare genetic variants within the terminal pathway of complement system in preeclampsia. Genes Immun 2025; 26:22-26. [PMID: 39690307 DOI: 10.1038/s41435-024-00310-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 10/11/2024] [Accepted: 11/22/2024] [Indexed: 12/19/2024]
Abstract
Preeclampsia is a common multifactorial disease of pregnancy. Dysregulation of complement activation is among emerging candidates responsible for disease pathogenesis. In a targeted exomic sequencing study of 609 women with preeclampsia and 2092 non-preeclamptic controls, we identified 14 variants within nine genes coding for components of the membrane attack complex (MAC, C5b-9) that are associated with preeclampsia. We found two rare missense variants in the C5 gene that predispose to preeclampsia (rs200674959: I1296V, OR (CI95) = 24.13 (1.25-467.43), p value = 0.01 and rs147430470: I330T, OR (CI95) = 22.75 (1.17-440.78), p value = 0.01). In addition, one predisposing rare variant and one protective rare variant were discovered in C6 (rs41271067: D396G, OR (CI95) = 2.93 (1.18-7.10), p value = 0.01 and rs114609505: T190I, 0.02 OR (CI95) = 0.47 (0.22-0.92), p value = 0.02). The results suggest that variants in the terminal complement pathway predispose to preeclampsia.
Collapse
Affiliation(s)
- A Inkeri Lokki
- Translational Immunology Research Program, Research Programs Unit and Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Heart and Lung Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland.
| | - Michael Triebwasser
- Department of Pediatrics, Division of Pediatric Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Emma Daly
- Hospital and Harvard Medical School, Boston, MA, USA
| | - Mitja I Kurki
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Markus Perola
- Department of Public Health and Welfare, National Institute for Health and Welfare, Helsinki, Finland
| | - Kirsi Auro
- Department of Public Health and Welfare, National Institute for Health and Welfare, Helsinki, Finland
| | - Jane E Salmon
- Hospital for Special Surgery; Weill Medical College of Cornell University, New York, NY, USA
| | - Java Anuja
- Division of Nephrology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Mark Daly
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - John P Atkinson
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannele Laivuori
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Obstetrics and Gynecology, Tampere University Hospital and the Wellbeing Services County of Pirkanmaa, Tampere, Finland
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Seppo Meri
- Translational Immunology Research Program, Research Programs Unit and Bacteriology and Immunology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- HUSLAB Diagnostic Center, Helsinki University Central Hospital, Helsinki, Finland.
| |
Collapse
|
3
|
Cecati M, Fumarola S, Vaiasicca S, Cianfruglia L, Vignini A, Giannubilo SR, Emanuelli M, Ciavattini A. Preeclampsia as a Study Model for Aging: The Klotho Gene Paradigm. Int J Mol Sci 2025; 26:902. [PMID: 39940672 PMCID: PMC11817256 DOI: 10.3390/ijms26030902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Aging and pregnancy are often considered opposites in a woman's biological timeline. Aging is defined by a gradual decline in the functional capabilities of an organism over its lifetime, while pregnancy is characterized by the presence of the transient placenta, which fosters the cellular fitness necessary to support fetal growth. However, in the context of preeclampsia, pregnancy and aging share common hallmarks, including clinical complications, altered cellular phenotypes, and heightened oxidative stress. Furthermore, women with pregnancies complicated by preeclampsia tend to experience age-related disorders earlier than those with healthy pregnancies. Klotho, a gene discovered fortuitously in 1997 by researchers studying aging mechanisms, is primarily expressed in the kidneys but also to a lesser extent in several other tissues, including the placenta. The Klotho protein is a membrane-bound protein that, upon cleavage by ADAM10/17, is released into the circulation as soluble Klotho (sKlotho) where it plays a role in modulating oxidative stress. This review focuses on the involvement of sKlotho in the development of preeclampsia and age-related disorders, as well as the expression of the recently discovered Mytho gene, which has been associated with skeletal muscle atrophy.
Collapse
Affiliation(s)
- Monia Cecati
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Stefania Fumarola
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (S.F.); (S.V.); (L.C.)
| | - Salvatore Vaiasicca
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (S.F.); (S.V.); (L.C.)
| | - Laura Cianfruglia
- Scientific Direction, IRCCS INRCA, 60124 Ancona, Italy; (S.F.); (S.V.); (L.C.)
| | - Arianna Vignini
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica Delle Marche, 60126 Ancona, Italy;
| | - Stefano Raffaele Giannubilo
- Department of Clinical Sciences, Clinic of Obstetrics and Gynaecology, Università Politecnica Delle Marche, 60123 Ancona, Italy;
| | - Monica Emanuelli
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Università Politecnica Delle Marche, 60126 Ancona, Italy;
| | - Andrea Ciavattini
- Department of Clinical Sciences, Clinic of Obstetrics and Gynaecology, Università Politecnica Delle Marche, 60123 Ancona, Italy;
| |
Collapse
|
4
|
Stenqvist A, Bungum M, Pinborg AB, Bogstad J, Englund AL, Grøndahl ML, Zedeler A, Hansson SR, Giwercman A. High sperm deoxyribonucleic acid fragmentation index is associated with an increased risk of preeclampsia following assisted reproduction treatment. Fertil Steril 2025; 123:97-104. [PMID: 39122087 DOI: 10.1016/j.fertnstert.2024.08.316] [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: 01/16/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE To study the association between sperm deoxyribonucleic acid fragmentation index (DFI) and the odds of preeclampsia and other adverse perinatal outcomes after in vitro fertilization (IVF) and intracytoplasmic sperm injection (ICSI) treatment. DESIGN A prospective cohort study including infertile couples undergoing conventional IVF or ICSI treatment and their children. Data regarding preeclampsia and perinatal outcomes were derived from the Swedish National Birth Register. PATIENT(S) A total of 1,594 infertile couples undergoing IVF or ICSI treatment and their 1,660 children conceived by assisted reproduction. EXPOSURE Sperm DFI measured by Sperm Chromatin Structure Assay. MAIN OUTCOME MEASURE(S) The primary outcome was preeclampsia. The secondary outcomes were preterm birth (PTB), low birth weight, low Apgar score, and small for gestational age. RESULT(S) With a DFI level of <20% as a reference, the odds ratio (OR) of preeclampsia statistically significantly increased in the group with a DFI level of ≥20% when IVF was used as the fertilization method (OR, 2.2; 95% confidence interval, 1.1-4.4). Already at the DFI levels of ≥10%, in IVF pregnancies, the OR of preeclampsia increased in a dose-response manner, from a prevalence of 3.1% in the reference group to >10% among those with a DFI level of ≥30%. The DFI was not associated with the OR of preeclampsia in the ICSI group. In the entire cohort, a DFI level of ≥20% was associated with an increased OR of PTB (OR, 1.4; 95% confidence interval, 1.0-2.0). CONCLUSION(S) High DFI level was associated with increased odds of PTB and, in IVF pregnancies, also increased odds of preeclampsia.
Collapse
Affiliation(s)
- Amelie Stenqvist
- Molecular Reproductive Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden; Reproductive Medicine Centre, Skåne University Hospital, Malmö, Sweden; Department of Obstetrics and Gynaecology, Skåne University Hospital, Malmö/Lund, Sweden.
| | - Mona Bungum
- Reproductive Medicine Centre, Skåne University Hospital, Malmö, Sweden
| | - Anja Bisgaard Pinborg
- Fertility Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Jeanette Bogstad
- Fertility Clinic, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | | | | | - Anne Zedeler
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Stefan R Hansson
- Department of Obstetrics and Gynaecology, Skåne University Hospital, Malmö/Lund, Sweden; Obstetrics and Gynaecology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Aleksander Giwercman
- Molecular Reproductive Medicine, Department of Translational Medicine, Lund University, Malmö, Sweden; Reproductive Medicine Centre, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
5
|
Ishikuro M, Obara T, Hasegawa M, Murakami K, Ueno F, Noda A, Onuma T, Matsuzaki F, Iwama N, Kikuya M, Sugawara J, Azegami T, Nakayama T, Mito A, Arata N, Metoki H, Kanda T, Kuriyama S. Subsequent high blood pressure and hypertension by hypertensive disorders of pregnancy: the Tohoku Medical Megabank Project Birth and Three-Generation Cohort Study. Hypertens Res 2025; 48:68-76. [PMID: 39394519 PMCID: PMC11700841 DOI: 10.1038/s41440-024-01936-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024]
Abstract
Hypertensive disorders of pregnancy can cause hypertension in the future. Understanding how the blood pressure values of women with and without hypertensive disorders of pregnancy differ will facilitate follow-up blood pressure monitoring in clinical settings. This study investigated the association between hypertensive disorders of pregnancy and subsequent high blood pressure and hypertension. This study used Japanese data from the Tohoku Medical Megabank Project Birth and Three-Generation Cohort Study. Follow-up systolic and diastolic blood pressures in normotensive women during pregnancy and those with hypertensive disorders of pregnancy were estimated using analysis of covariance adjusted for women with low birthweight, history of gestational diabetes mellitus, age, body mass index, pulse rate, smoking and drinking at the follow-up assessment, paternal hypertension history, and maternal hypertension or hypertensive disorders of pregnancy history. Finally, we performed a multiple logistic regression analysis. In total, 7343 women were included in the analysis. Women with a history of hypertensive disorders of pregnancy had higher blood pressure approximately three years postpartum compared with normotensive women. Hypertensive disorders of pregnancy in the most recent pregnancy in different subgroups, such as nulliparous women, multiparous women without a history of hypertensive disorders of pregnancy, and multiparous women with a history of hypertensive disorders of pregnancy, were associated with an increased risk of subsequent hypertension. Women's birthweight was also weakly associated with hypertension. Even one experience of hypertensive disorders of pregnancy may contribute to elevated blood pressure and hypertension approximately three years postpartum. In addition, women's birthweights may have a weak relationship with increasing blood pressure.
Collapse
Affiliation(s)
- Mami Ishikuro
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan.
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.
| | - Taku Obara
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Mayu Hasegawa
- Tohoku University School of Medicine, Sendai, Miyagi, Japan
| | - Keiko Murakami
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Fumihiko Ueno
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Aoi Noda
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Tomomi Onuma
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Fumiko Matsuzaki
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Noriyuki Iwama
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Masahiro Kikuya
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Teikyo University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Junichi Sugawara
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- Tohoku University Hospital, Sendai, Miyagi, Japan
| | - Tatsuhiko Azegami
- Department of Internal Medicine, Keio University School of Medicine, Shinjyuku-ku, Tokyo, Japan
| | - Takashin Nakayama
- Department of Internal Medicine, Keio University School of Medicine, Shinjyuku-ku, Tokyo, Japan
| | - Asako Mito
- Division of Maternal Medicine, Center for Maternal-Fetal, Neonatal, and Reproductive Medicine, National Center for Child Health and Development, Setagaya, Japan
| | - Naoko Arata
- Division of Maternal Medicine, Center for Maternal-Fetal, Neonatal, and Reproductive Medicine, National Center for Child Health and Development, Setagaya, Japan
| | - Hirohito Metoki
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Takeshi Kanda
- Department of Nephrology, Faculty of Medicine, Shimane University, Izumo, Shimane, Japan
| | - Shinichi Kuriyama
- Department of Preventive Medicine and Epidemiology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
- International Research Institute of Disaster Science, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
6
|
Sergent F, Vaiman D, Raia‐Barjat T, Younes H, Marquette C, Desseux M, Nahed RA, Kieu T, Dung NV, Keck M, Hoffmann P, Murthi P, Benharouga M, Alfaidy N. Antagonisation of Prokineticin Receptor-2 Attenuates Preeclampsia Symptoms. J Cell Mol Med 2025; 29:e70346. [PMID: 39817714 PMCID: PMC11736873 DOI: 10.1111/jcmm.70346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/18/2025] Open
Abstract
Preeclampsia (PE) is the most threatening pathology of human pregnancy. Placenta from PE patients releases harmful factors that contribute to the exacerbation of the disease. Among these factors is the prokineticin1 (PROK1) and its receptor, PROKR2 that we identified as a mediators of PE. Here we tested the effects of PKRA, an antagonist of PROKR2, on the attenuation of PE symptoms. We used the genetic PE mouse model, STOX1 that overexpresses Stox1 gene in a heterozygosis manner in the placenta. This model allowed exploiting two genotypes of the offspring, those that overexpress the Stox1 gene, and the WT that grow in a PE environment (STE). We characterised the effect PKRA (1 μM) on the attenuation of PE symptoms and compared its effects on STOX1 and STE placentas. We also used STOX1 overexpressing trophoblast cells to decipher the PROK1-underlying mechanism. We demonstrated that (i) antagonisation of PROKR2 attenuated PE-mediated hypertension and proteinuria, (ii) STE placentas and foetuses exhibited better outcomes in response to PKRA, (iii) the secretome of STOX1-trophoblasts impacted the integrity of the fetal vasculature that was attenuated by PKRA treatment. This study demonstrates the direct involvement of the PROK1 in PE and identifies PKRA as a promising therapy for PE.
Collapse
Affiliation(s)
- Frédéric Sergent
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Daniel Vaiman
- Institute Cochin, U1016, INSERM, UMR 8504 CNRS, Paris‐Descartes UniversitéParisFrance
| | - Tiphaine Raia‐Barjat
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Hadi Younes
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Christel Marquette
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Morgane Desseux
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Roland Abi Nahed
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Trinh‐Le‐Vi Kieu
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Nguyen Viet Dung
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Mathilde Keck
- Université Paris Saclay, CEA, INRAE, Département Médicaments et Technologies Pour la Santé (DMTS)Gif‐sur‐YvetteFrance
| | - Pascale Hoffmann
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
- Centre Hospitalo‐Universitaire Grenoble Alpes, Service Obstétrique, CS 10217GrenobleFrance
- Université Grenoble AlpesGrenobleFrance
| | - Padma Murthi
- Department of PharmacologyMonash Biomedicine Discovery Institute, Monash UniversityMelbourneVictoriaAustralia
- Department of Maternal‐Fetal Medicine Pregnancy Research CentreThe Royal Women's HospitalMelbourneVictoriaAustralia
- Department of Obstetrics and GynecologyThe University of MelbourneMelbourneVictoriaAustralia
| | - Mohamed Benharouga
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
| | - Nadia Alfaidy
- Interdisciplinary Research Institute of Grenoble, IRIG‐BiosantéUniversity Grenoble Alpes, INSERM, CEA, UMR 1292GrenobleFrance
- Commissariat à l'Energie Atomique et Aux Energies Alternatives (CEA)Biosciences and Biotechnology Institute of GrenobleGrenobleFrance
- Centre Hospitalo‐Universitaire Grenoble Alpes, Service Obstétrique, CS 10217GrenobleFrance
- Université Grenoble AlpesGrenobleFrance
| |
Collapse
|
7
|
Shenhav S, Harel I, Solt I, Shenhav A, Fytlovich S, Aharoni D, Rimler A, Anteby EY, Ovadia YS. Fetoplacental unit involvement in uric acid production in women with severe preeclampsia: a prospective case control pilot study. J Matern Fetal Neonatal Med 2024; 37:2399304. [PMID: 39287009 DOI: 10.1080/14767058.2024.2399304] [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: 04/18/2024] [Revised: 08/08/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE Preeclampsia (PE) is a common complication of pregnancy that carries significant risks for both the mother and the fetus, and is frequently accompanied by hyperuricemia, yet the exact source of elevated uric acid (UA) levels remains partially elucidated. Several potential origins for increased UA levels include abnormal renal function, increased tissue breakdown, and increased activity of the enzyme Xanthine Oxidase (XO). The aim of the study was to determine serum levels of UA and XO not only in maternal serum, but also in umbilical vein (UV) and umbilical artery (UA) and explore their possible role in PE development. METHODS A prospective case-control pilot study was conducted in women who were found positive for PE with severe features, and had elevated UA levels above 6 mg/dL, with normotensive pregnant women serving as controls. Renal function, UA and XO levels were measured in maternal, UV and UA serums immediately after delivery. They were then compared between PE (n = 21) and control (n = 18) groups, as well as across all mediums (maternal, UV and UA) among the total study sample (N = 39). Diastolic blood pressure (DBP) was also measured immediately following delivery. RESULTS The mean serum maternal creatinine levels did not differ significantly between groups (0.65 ± 0.03 vs 0.6 ± 0.07, p = 0.13). Both mean maternal serum UA and XO concentrations were higher in PE group than in control (7.3 ± 1.2 vs 4.2 ± 0.9, p < 0.01 and 3.6 ± 3.5 Vs 1.7 ± 0.8, p < 0.01, respectively). The mean UV and UA serum XO concentrations were significantly higher in PE group compared to control (4.2 ± 3.6 vs 2.2 ± 1.4, p < 0.01 and 4.2 ± 3.6 vs 2.1 ± 1.5, p < 0.01, respectively). Polynomial fit correlation test demonstrated a significant association between maternal DBP and UV XO concentration for all the total study participants (p = 0.03). CONCLUSION Despite preserved renal functions, UA and XO levels were elevated in women with PE. Importantly, this pattern was found to be applied to the feto-placental unit as well, which may indicate an active involvement of the fetus in the hypoxic process. Further study is needed to clarify the possible role of the feto-placental unit in pregnancies complicated by PE.
Collapse
Affiliation(s)
- Simon Shenhav
- Obstetrics and Gynecology Division, Barzilai University Medical Center Ashkelon, Ashkelon, Israel
- Faculty of Health Sciences, Ben-Gurion University of Negev, Ashkelon, Israel
| | - Iris Harel
- Obstetrics and Gynecology Division, Barzilai University Medical Center Ashkelon, Ashkelon, Israel
| | - Ido Solt
- Department of Obstetrics and Gynecology, Rambam Health Care Campus, Haifa, Israel
| | - Amit Shenhav
- Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Shlomo Fytlovich
- Laboratory of Clinical Biochemistry, Barzilai University Medical Center Ashkelon, Ashkelon, Israel
| | - Dorit Aharoni
- Laboratory of Clinical Biochemistry, Barzilai University Medical Center Ashkelon, Ashkelon, Israel
| | - Avi Rimler
- Laboratory of Clinical Biochemistry, Barzilai University Medical Center Ashkelon, Ashkelon, Israel
| | - Eyal Y Anteby
- Obstetrics and Gynecology Division, Barzilai University Medical Center Ashkelon, Ashkelon, Israel
- Faculty of Health Sciences, Ben-Gurion University of Negev, Ashkelon, Israel
| | - Yaniv S Ovadia
- Obstetrics and Gynecology Division, Barzilai University Medical Center Ashkelon, Ashkelon, Israel
| |
Collapse
|
8
|
Feng W, Luo Y. Preeclampsia and its prediction: traditional versus contemporary predictive methods. J Matern Fetal Neonatal Med 2024; 37:2388171. [PMID: 39107137 DOI: 10.1080/14767058.2024.2388171] [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: 04/22/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVE Preeclampsia (PE) poses a significant threat to maternal and perinatal health, so its early prediction, prevention, and management are of paramount importance to mitigate adverse pregnancy outcomes. This article provides a brief review spanning epidemiology, etiology, pathophysiology, and risk factors associated with PE, mainly discussing the emerging role of Artificial Intelligence (AI) deep learning (DL) technology in predicting PE, to advance the understanding of PE and foster the clinical application of early prediction methods. METHODS Our narrative review comprehensively examines the PE epidemiology, etiology, pathophysiology, risk factors and predictive approaches, including traditional models and AI deep learning technology. RESULTS Preeclampsia involves a wide range of biological and biochemical risk factors, among which poor uterine artery remodeling, excessive immune response, endothelial dysfunction, and imbalanced angiogenesis play important roles. Traditional PE prediction models exhibit significant limitations in sensitivity and specificity, particularly in predicting late-onset PE, with detection rates ranging from only 30% to 50%. AI models have exhibited a notable level of predictive accuracy and value across various populations and datasets, achieving detection rates of approximately 70%. Particularly, they have shown superior predictive capabilities for late-onset PE, thereby presenting novel opportunities for early screening and management of the condition. CONCLUSION AI DL technology holds promise in revolutionizing the prediction and management of PE. AI-based approaches offer a pathway toward more effective risk assessment methods by addressing the shortcomings of traditional prediction models. Ongoing research efforts should focus on expanding databases and validating the performance of AI in diverse populations, leading to the development of more sophisticated prediction models with improved accuracy.
Collapse
Affiliation(s)
- Wei Feng
- Department of Gynecology, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| | - Ying Luo
- Department of Gynecology, China Aerospace Science & Industry Corporation 731 Hospital, Beijing, China
| |
Collapse
|
9
|
Nurkkala J, Vaura F, Toivonen J, Niiranen T. Genetics of hypertension-related sex differences and hypertensive disorders of pregnancy. Blood Press 2024; 33:2408574. [PMID: 39371034 DOI: 10.1080/08037051.2024.2408574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024]
Abstract
Background: Hypertension and hypertensive disorders of pregnancy (HDP) cause a significant burden of disease on societies and individuals by increasing cardiovascular disease risk. Environmental risk factors alone do not explain the observed sexual dimorphism in lifetime blood pressure (BP) trajectories nor inter-individual variation in HDP risk. Methods: In this short review, we focus on the genetics of hypertension-related sex differences and HDP and discuss the importance of genetics utilization for sex-specific hypertension risk prediction. Results: Population and twin studies estimate that 28-66% of variation in BP levels and HDP is explained by genetic variation, while genomic wide association studies suggest that BP traits and HDP partly share a common genetic background. Moreover, environmental and epigenetic regulation of these genes differ by sex and oestrogen receptors in particular are shown to convey cardio- and vasculoprotective effects through epigenetic regulation of DNA. The majority of known genetic variation in hypertension and HDP is polygenic. Polygenic risk scores for BP display stronger associations with hypertension risk in women than in men and are associated with sex-specific age of hypertension onset. Monogenic forms of hypertension are rare and mostly present equally in both sexes. Conclusion: Despite recent genetic discoveries providing new insights into HDP and sex differences in BP traits, further research is needed to elucidate the underlying biology. Emphasis should be placed on demonstrating the added clinical value of these genetic discoveries, which may eventually facilitate genomics-based personalized treatments for hypertension and HDP.
Collapse
Affiliation(s)
- Jouko Nurkkala
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
- Department of Anesthesiology and Intensive Care, University of Turku, Turku, Finland
| | - Felix Vaura
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Jenni Toivonen
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital, Turku, Finland
- Department of Anesthesiology and Intensive Care, University of Turku, Turku, Finland
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Turku, Finland
- Division of Medicine, Turku University Hospital, Turku, Finland
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Turku, Finland
| |
Collapse
|
10
|
Smith J, Powell M, Cromartie W, Smith S, Jones K, Castillo A, Shaw J, Editone J, Howard A, Tatum R, Smith A, Fisher B, Booz GW, Cunningham M. Intrauterine growth-restricted pregnant rats, from placental ischemic dams, display preeclamptic-like symptoms: A new rat model of preeclampsia. Physiol Rep 2024; 12:e70112. [PMID: 39482843 PMCID: PMC11527824 DOI: 10.14814/phy2.70112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024] Open
Abstract
Preeclampsia (PE) is characterized by de novo hypertension (HTN) and is often associated with intrauterine growth restriction (IUGR). Hallmarks of PE are placental ischemia, decreased nitric oxide (NO) bioavailability, oxidative stress (OS), and organ damage in the kidneys and brain. This study aims to characterize a new model of PE using pregnant IUGR rats from hypertensive placental ischemic dams. It is hypothesized that pregnant IUGR rats from hypertensive placental ischemic dams will have elevated blood pressure (BP), OS, and organ damage. In this study, pregnant rats are divided into two groups: normal pregnant (NP) and hypertensive placental ischemic dams (RUPP). Offspring from NP and RUPP dams were mated at 10 weeks of age to generate pregnant IUGR (IUGR Preg) and pregnant control (CON Preg) rats. BP and other markers of PE were evaluated during late gestation. Pregnant IUGR rats had elevated BP and systemic OS. The maternal body weight of pregnant IUGR rats and their pups' weights were decreased, while the brains were enlarged with elevated OS. In summary, pregnant IUGR rats, born from hypertensive placental ischemic dams, have HTN and increased systemic and brain OS, with larger brain sizes and smaller pups. Furthermore, this study shows that pregnant IUGR rats exhibit a preeclamptic-like phenotype, suggesting a new epigenetic model of PE.
Collapse
Affiliation(s)
- Jonna Smith
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Madison Powell
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Whitney Cromartie
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Savanna Smith
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Kylie Jones
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Angie Castillo
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Jordan Shaw
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Joseph Editone
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| | - Ahfiya Howard
- School of Social WorkTexas A & M University‐CommerceCommerceTexasUSA
| | - Robert Tatum
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Alex Smith
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Brandon Fisher
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - George W. Booz
- Department of Pharmacology & ToxicologyUniversity of Mississippi Medical CenterJacksonMississippiUSA
| | - Mark Cunningham
- Department of Physiology and AnatomyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| |
Collapse
|
11
|
Wu Y, Sun T, Medina P, Narasimhan P, Stevenson DK, Von Versen-Höynck F, Armstrong J, Wu JC, Sayed N, Winn VD. A Novel Stem Cell Model to Study Preeclampsia Endothelial Dysfunction. Reprod Sci 2024; 31:2993-3003. [PMID: 39179924 PMCID: PMC11438721 DOI: 10.1007/s43032-024-01590-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/08/2024] [Indexed: 08/26/2024]
Abstract
Preeclampsia is a common pregnancy complication affecting 5% to 7% of all pregnancies worldwide annually. While the pathogenesis is not fully understood, maternal endothelium dysfunction is thought to be a central component to preeclampsia development. Studies to dissect maternal endothelial dysfunction, particularly on a patient-specific basis, are hampered by limited access to systemic primary endothelial cells (ECs). The objective of this study was to establish a replenishable, patient-specific in vitro EC model to allow robust mechanistic studies to dissect endothelial dysfunction in preeclampsia. Induced pluripotent stem cells (iPSCs) from three women with a history of normotensive pregnancies were differentiated into ECs. The established ECs were exposed to pooled sera from normotensive pregnancies, preeclamptic pregnancies, normotensive postpartum for non-pregnant comparison and controls. Endothelial functions including nitric oxide (NO) release, cell migration, tube formation and viability were evaluated. Levels of NO release were significantly lower after incubation with preeclamptic sera compared to the fetal bovine serum (FBS) control, and normotensive and non-pregnant (postpartum) sera treatments were also lower than FBS but higher than preeclamptic sera treatments. Tube formation and cell migration were also impaired with preeclamptic sera compared to FBS controls. Cell viabilities remained unaffected by any sera treatment. Consistent outcomes were obtained across all three patient-specific lines treated with the same pooled sera. Establishment of patient-derived iPSC-ECs treated with pregnancy sera serves as a novel model to explore the interplay between individual maternal endothelial health and circulating factors that lead to endothelial dysfunction in preeclampsia.
Collapse
Affiliation(s)
- Yanming Wu
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Tianyanxin Sun
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - Pedro Medina
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Purnima Narasimhan
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA
| | - David K Stevenson
- Department of Pediatrics, Division of Neonatal and Developmental Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Frauke Von Versen-Höynck
- Department of Obstetrics, Gynecology and Reproductive Sciences, Hannover Medical School, Hannover, Germany
| | - Jennifer Armstrong
- Department of Pediatrics, Section of Neurology and Department of Obstetrics and Gynecology, Division of Basic Reproductive Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Vascular Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
12
|
Cao C, Saxena R, Gray KJ. Placental Origins of Preeclampsia: Insights from Multi-Omic Studies. Int J Mol Sci 2024; 25:9343. [PMID: 39273292 PMCID: PMC11395466 DOI: 10.3390/ijms25179343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Preeclampsia (PE) is a major cause of maternal and neonatal morbidity and mortality worldwide, with the placenta playing a central role in disease pathophysiology. This review synthesizes recent advancements in understanding the molecular mechanisms underlying PE, focusing on placental genes, proteins, and genetic variants identified through multi-omic approaches. Transcriptomic studies in bulk placental tissue have identified many dysregulated genes in the PE placenta, including the PE signature gene, Fms-like tyrosine kinase 1 (FLT1). Emerging single-cell level transcriptomic data have revealed key cell types and molecular signatures implicated in placental dysfunction and PE. However, the considerable variability among studies underscores the need for standardized methodologies and larger sample sizes to enhance the reproducibility of results. Proteomic profiling of PE placentas has identified numerous PE-associated proteins, offering insights into potential biomarkers and pathways implicated in PE pathogenesis. Despite significant progress, challenges such as inconsistencies in study findings and lack of validation persist. Recent fetal genome-wide association studies have identified multiple genetic loci associated with PE, with ongoing efforts to elucidate their impact on placental gene expression and function. Future directions include the integration of multi-omic data, validation of findings in diverse PE populations and clinical subtypes, and the development of analytical approaches and experimental models to study the complex interplay of placental and maternal factors in PE etiology. These insights hold promise for improving risk prediction, diagnosis, and management of PE, ultimately reducing its burden on maternal and neonatal health.
Collapse
Affiliation(s)
- Chang Cao
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Richa Saxena
- Center for Genomic Medicine and Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Kathryn J. Gray
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
13
|
Manoharan A, Ballambattu VB, Palani R. Genetic architecture of preeclampsia. Clin Chim Acta 2024; 558:119656. [PMID: 38583550 DOI: 10.1016/j.cca.2024.119656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/04/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Affiliation(s)
- Aarthi Manoharan
- Department of Medical Biotechnology, Kirumampakkam, Puducherry 607403, India.
| | | | - Ramya Palani
- Department of Obstetrics and Gynecology, Aarupadai Veedu Medical College and Hospital, Vinayaka Mission's Research Foundation (DU), Kirumampakkam, Puducherry 607403, India
| |
Collapse
|
14
|
Gong X, He W, Jin W, Ma H, Wang G, Li J, Xiao Y, Zhao Y, Chen Q, Guo H, Yang J, Qi Y, Dong W, Fu M, Li X, Liu J, Liu X, Yin A, Zhang Y, Wei Y. Disruption of maternal vascular remodeling by a fetal endoretrovirus-derived gene in preeclampsia. Genome Biol 2024; 25:117. [PMID: 38715110 PMCID: PMC11075363 DOI: 10.1186/s13059-024-03265-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Preeclampsia, one of the most lethal pregnancy-related diseases, is associated with the disruption of uterine spiral artery remodeling during placentation. However, the early molecular events leading to preeclampsia remain unknown. RESULTS By analyzing placentas from preeclampsia, non-preeclampsia, and twin pregnancies with selective intrauterine growth restriction, we show that the pathogenesis of preeclampsia is attributed to immature trophoblast and maldeveloped endothelial cells. Delayed epigenetic reprogramming during early extraembryonic tissue development leads to generation of excessive immature trophoblast cells. We find reduction of de novo DNA methylation in these trophoblast cells results in selective overexpression of maternally imprinted genes, including the endoretrovirus-derived gene PEG10 (paternally expressed gene 10). PEG10 forms virus-like particles, which are transferred from the trophoblast to the closely proximate endothelial cells. In normal pregnancy, only a low amount of PEG10 is transferred to maternal cells; however, in preeclampsia, excessive PEG10 disrupts maternal vascular development by inhibiting TGF-beta signaling. CONCLUSIONS Our study reveals the intricate epigenetic mechanisms that regulate trans-generational genetic conflict and ultimately ensure proper maternal-fetal interface formation.
Collapse
Affiliation(s)
- Xiaoli Gong
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Wei He
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wan Jin
- Euler Technology, Beijing, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongwei Ma
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China
- Department Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Gang Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
- Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiaxin Li
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yu Xiao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Human Genetic Resources Preservation Center of Hubei Province, Wuhan, China
- Laboratory of Precision Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | | | | | - Jiexia Yang
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Yiming Qi
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Wei Dong
- Maternity Ward, Haidian Maternal and Child Health Hospital, Beijing, China
| | - Meng Fu
- Department of Obstetrics and Gynecology, Haidian Maternal and Child Health Hospital, Beijing, China
| | - Xiaojuan Li
- Euler Technology, Beijing, China
- Present Address: International Max Planck Research School for Genome Science, and University of Göttingen, Göttingen Center for Molecular Biosciences, Göttingen, Germany
| | | | - Xinghui Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, China.
- Department Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China.
| | - Aihua Yin
- Medical Genetic Center, Guangdong Women and Children Hospital, Guangzhou, China.
| | - Yi Zhang
- Euler Technology, Beijing, China.
| | - Yuan Wei
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
15
|
Lokki A, Triebwasser M, Daly E, Cohort F, Kurki M, Perola M, Auro K, Salmon J, Java A, Daly M, Atkinson J, Laivuori H, Meri S. Rare variants in genes coding for components of the terminal pathway of the complement system in preeclampsia. RESEARCH SQUARE 2024:rs.3.rs-4121735. [PMID: 38645143 PMCID: PMC11030519 DOI: 10.21203/rs.3.rs-4121735/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Preeclampsia is a common multifactorial disease of pregnancy. Dysregulation of the complement activation is among emerging candidates responsible for disease pathogenesis. In a targeted exomic sequencing study we identified 14 variants within nine genes coding for components of the membrane attack complex (MAC, C5b-9) that are associated with preeclampsia. We found two rare missense variants in the C5 gene that predispose to preeclampsia (rs200674959: I1296V, OR (CI95) = 24.13 (1.25-467.43), p-value = 0.01 and rs147430470: I330T, OR (CI95) = 22.75 (1.17-440.78), p-value = 0.01). In addition, one predisposing rare variant and one protective rare variant were discovered in C6 (rs41271067: D396G, OR (CI95) = 2.93 (1.18-7.10), p-value = 0.01 and rs114609505: T190I, 0.02 OR (CI95) = 0.47 (0.22-0.92), p-value = 0.02). The results suggest that variants in terminal complement pathway predispose to preeclampsia.
Collapse
|
16
|
Costa L, Bermudez-Guzman L, Benouda I, Laissue P, Morel A, Jiménez KM, Fournier T, Stouvenel L, Méhats C, Miralles F, Vaiman D. Linking genotype to trophoblast phenotype in preeclampsia and HELLP syndrome associated with STOX1 genetic variants. iScience 2024; 27:109260. [PMID: 38439971 PMCID: PMC10910284 DOI: 10.1016/j.isci.2024.109260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/20/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
Preeclampsia is a major hypertensive pregnancy disorder with a 50% heritability. The first identified gene involved in the disease is STOX1, a transcription factor, whose variant Y153H predisposes to the disease. Two rare mutations were also identified in Colombian women affected by the hemolysis, elevated liver enzyme, low platelet syndrome, a complication of preeclampsia (T188N and R364X). Here, we explore the effects of these variants in trophoblast cell models (BeWo) where STOX1 was previously invalidated. We firstly showed that STOX1 knockout alters response to oxidative stress, cell proliferation, and fusion capacity. Then, we showed that mutant versions of STOX1 trigger alterations in gene profiles, growth, fusion, and oxidative stress management. The results also reveal alterations of the STOX interaction with DNA when the mutations affected the DNA-binding domain of STOX1 (Y153H and T188N). We also reveal here that a major contributor of these effects appears to be the E2F3 transcription factor.
Collapse
Affiliation(s)
- Lorenzo Costa
- Institut Cochin, Team ‘From Gametes To Birth’, INSERM U1016, CNRS UMR8104, Université de Paris, 24 rue du Faubourg St Jacques, 75014 Paris, France
- Department of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | | | - Ikram Benouda
- Institut Cochin, Team ‘From Gametes To Birth’, INSERM U1016, CNRS UMR8104, Université de Paris, 24 rue du Faubourg St Jacques, 75014 Paris, France
| | - Paul Laissue
- Biopas Laboratoires, Orphan Diseases Unit, BIOPAS GROUP, Bogotá 111111, Colombia
| | - Adrien Morel
- Universidad Del Rosario, School of Medicine and Health Sciences, Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), Bogotá, Colombia
| | - Karen Marcela Jiménez
- Universidad Del Rosario, School of Medicine and Health Sciences, Center for Research in Genetics and Genomics (CIGGUR), Institute of Translational Medicine (IMT), Bogotá, Colombia
| | - Thierry Fournier
- Université Paris Cité, INSERM, UMR-S1139, Pathophysiology & Pharmacotoxicology of the Human Placenta, Pre- & Post-natal Microbiota (3PHM), 75006 Paris, France
| | - Laurence Stouvenel
- Institut Cochin, Team ‘From Gametes To Birth’, INSERM U1016, CNRS UMR8104, Université de Paris, 24 rue du Faubourg St Jacques, 75014 Paris, France
| | - Céline Méhats
- Institut Cochin, Team ‘From Gametes To Birth’, INSERM U1016, CNRS UMR8104, Université de Paris, 24 rue du Faubourg St Jacques, 75014 Paris, France
| | - Francisco Miralles
- Institut Cochin, Team ‘From Gametes To Birth’, INSERM U1016, CNRS UMR8104, Université de Paris, 24 rue du Faubourg St Jacques, 75014 Paris, France
| | - Daniel Vaiman
- Institut Cochin, Team ‘From Gametes To Birth’, INSERM U1016, CNRS UMR8104, Université de Paris, 24 rue du Faubourg St Jacques, 75014 Paris, France
| |
Collapse
|
17
|
Frimat M, Gnemmi V, Stichelbout M, Provôt F, Fakhouri F. Pregnancy as a susceptible state for thrombotic microangiopathies. Front Med (Lausanne) 2024; 11:1343060. [PMID: 38476448 PMCID: PMC10927739 DOI: 10.3389/fmed.2024.1343060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/12/2024] [Indexed: 03/14/2024] Open
Abstract
Pregnancy and the postpartum period represent phases of heightened vulnerability to thrombotic microangiopathies (TMAs), as evidenced by distinct patterns of pregnancy-specific TMAs (e.g., preeclampsia, HELLP syndrome), as well as a higher incidence of nonspecific TMAs, such as thrombotic thrombocytopenic purpura or hemolytic uremic syndrome, during pregnancy. Significant strides have been taken in understanding the underlying mechanisms of these disorders in the past 40 years. This progress has involved the identification of pivotal factors contributing to TMAs, such as the complement system, ADAMTS13, and the soluble VEGF receptor Flt1. Regardless of the specific causal factor (which is not generally unique in relation to the usual multifactorial origin of TMAs), the endothelial cell stands as a central player in the pathophysiology of TMAs. Pregnancy has a major impact on the physiology of the endothelium. Besides to the development of placenta and its vascular consequences, pregnancy modifies the characteristics of the women's microvascular endothelium and tends to render it more prone to thrombosis. This review aims to delineate the distinct features of pregnancy-related TMAs and explore the contributing mechanisms that lead to this increased susceptibility, particularly influenced by the "gravid endothelium." Furthermore, we will discuss the potential contribution of histopathological studies in facilitating the etiological diagnosis of pregnancy-related TMAs.
Collapse
Affiliation(s)
- Marie Frimat
- CHU Lille, Nephrology Department, Univ. Lille, Lille, France
- Inserm, Institut Pasteur de Lille, Univ. Lille, Lille, France
| | | | | | - François Provôt
- CHU Lille, Nephrology Department, Univ. Lille, Lille, France
| | - Fadi Fakhouri
- Service of Nephrology and Hypertension, CHUV and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
18
|
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, Commodore-Mensah Y, Currie ME, Elkind MSV, Evenson KR, Generoso G, Heard DG, Hiremath S, Johansen MC, Kalani R, Kazi DS, Ko D, Liu J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Perman SM, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Tsao CW, Urbut SM, Van Spall HGC, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024; 149:e347-e913. [PMID: 38264914 DOI: 10.1161/cir.0000000000001209] [Citation(s) in RCA: 488] [Impact Index Per Article: 488.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2024 AHA Statistical Update is the product of a full year's worth of effort in 2023 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. The AHA strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional global data, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
19
|
Hughes ZH, Hughes LM, Khan SS. Genetic contributions to risk of adverse pregnancy outcomes. CURRENT CARDIOVASCULAR RISK REPORTS 2023; 17:185-193. [PMID: 38186860 PMCID: PMC10768680 DOI: 10.1007/s12170-023-00729-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2023] [Indexed: 01/09/2024]
Abstract
Purpose of Review Adverse pregnancy outcomes (APOs), including hypertensive disorders of pregnancy (HDP), low birthweight (LBW), and preterm birth (PTB), along with peripartum cardiomyopathy (PPCM) are associated with short- and long-term maternal and fetal cardiovascular risks. This review focuses on the genetic contributions to the risk of APOs and PPCM. Recent Findings The expansion of genome-wide association studies (GWAS) has led to better understanding of the biologic mechanisms underpinning APO, PPCM, and the predisposition to cardiovascular disease across the life course. Genetic loci known to be involved with the risk of hypertension (FTO, ZNF831) have been associated with the development of overall HDP and preeclampsia. Additionally, four loci significantly associated with type 2 diabetes have been associated with GDM (CDKAL1, MTNR1B, TCF7L2, CDK2NA-CDKN2B). Variants in loci known to affect genes coding for proteins involved in immune cell function and placental health (EBF1, EEFSEC, AGTR2, 2q13) have been implicated in the development of PTB and future cardiovascular risks for both the mother and the offspring. Genetic similarities in rare variants between PPCM and dilated cardiomyopathy have been described suggesting shared pathophysiologic origins as well as predisposition for future risk of heart failure, highlighting the need for the development PPCM genetic counseling guidelines. Summary Genetics may inform mechanisms, risk, and counseling for individuals after an APO or PPCM. Through recent advances in genetic techniques and analytic approaches, new insights into the underlying biologic mechanisms and genetic variants leading to these risks have been discovered.
Collapse
Affiliation(s)
- Zachary H. Hughes
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, UA
| | - Lydia M. Hughes
- Department of Obstetrics & Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, UA
| | - Sadiya S. Khan
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Division of Cardiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
20
|
Huluta I, Wright A, Cosma LM, Hamed K, Nicolaides KH, Charakida M. Fetal Cardiac Function at Midgestation and Subsequent Development of Preeclampsia. J Am Soc Echocardiogr 2023; 36:1110-1115. [PMID: 37230422 DOI: 10.1016/j.echo.2023.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE To assess differences in cardiac morphology and function at midgestation in fetuses from pregnancies that subsequently developed preeclampsia (PE) or gestational hypertension (GH). METHODS This was a prospective study in 5,801 women with singleton pregnancies attending for a routine ultrasound examination at midgestation, including 179 (3.1%) who subsequently developed PE and 149 (2.6%) who developed GH. Conventional and more advanced echocardiographic modalities, such as speckle-tracking, were used to assess fetal cardiac function in the right and left ventricle. The morphology of the fetal heart was assessed by calculating the right and left sphericity index. RESULTS In fetuses from the PE group (vs the no PE or GH group) there was a significantly higher left ventricular global longitudinal strain and lower left ventricular ejection fraction that could not be accounted for by fetal size. All other indices of fetal cardiac morphology and function were comparable between groups. There was no significant correlation between fetal cardiac indices and uterine artery pulsatility index multiple of the median or placental growth factor multiple of the median. CONCLUSION At midgestation, fetuses of mothers at risk of developing PE, but not those at risk of GH, have mild reduction in left ventricular myocardial function. Although absolute differences were minimal and most likely not clinically relevant, these may suggest an early programming effect on left ventricular contractility in fetuses of mothers who develop PE.
Collapse
Affiliation(s)
- Iulia Huluta
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Alan Wright
- Institute of Health Research, University of Exeter, Exeter, United Kingdom
| | - Livia Mihaela Cosma
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Karam Hamed
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Marietta Charakida
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom; School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom.
| |
Collapse
|
21
|
Chen Y, Huang X, Wu S, Guo P, Huang J, Zhou L, Tan X. Machine-learning predictive model of pregnancy-induced hypertension in the first trimester. Hypertens Res 2023; 46:2135-2144. [PMID: 37160966 DOI: 10.1038/s41440-023-01298-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/17/2023] [Accepted: 03/17/2023] [Indexed: 05/11/2023]
Abstract
In the first trimester of pregnancy, accurately predicting the occurrence of pregnancy-induced hypertension (PIH) is important for both identifying high-risk women and adopting early intervention. In this study, we used four machine-learning models (LASSO logistic regression, random forest, backpropagation neural network, and support vector machines) to predict the occurrence of PIH in a prospective cohort. Candidate features for predicting the occurrence of middle and late PIH were acquired using a LASSO algorithm. The performance of predictive models was assessed using receiver operating characteristic analysis. Finally, a nomogram was established with the model scores, age, and nulliparity. Calibration, clinical usefulness, and internal validation were used to assess the performance of the nomogram. In the training set (2258 pregnant women), eleven candidate factors in the first trimester were significantly associated with the occurrence of PIH (P < 0.001 in the training set). Four models showed AUCs from 0.780 to 0.816 in the training set. For the validation set (939 pregnant women), AUCs varied from 0.516 to 0.795. The nomogram showed good discrimination, with an AUC of 0.847 (95% CI: 0.805-0.889) in the training set and 0.753 (95% CI: 0.653-0.853) in the validation set. Decision curve analysis suggested that the model was clinically useful. The model developed using LASSO logistic regression achieved the best performance in predicting the occurrence of PIH. The derived nomogram, which incorporates the model score and maternal risk factors, can be used to predict PIH in clinical practice. We develop a model with good performance for clinical prediction of PIH in the first trimester.
Collapse
Affiliation(s)
- Yequn Chen
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Xiru Huang
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
- Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Shiwan Wu
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Pi Guo
- Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Ju Huang
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Li Zhou
- Cancer Hospital Of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Xuerui Tan
- The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China.
- Shantou University Medical College, Shantou, Guangdong, 515041, China.
| |
Collapse
|
22
|
Khezri R, Salarilak S, Jahanian S. The association between maternal anemia during pregnancy and preterm birth. Clin Nutr ESPEN 2023; 56:13-17. [PMID: 37344062 DOI: 10.1016/j.clnesp.2023.05.003] [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: 09/08/2022] [Revised: 04/25/2023] [Accepted: 05/03/2023] [Indexed: 06/23/2023]
Abstract
BACKGROUND & AIMS Maternal anemia is one of the most serious health problems during pregnancy. The causes of anemia include iron deficiency, parasitic diseases, micronutrient deficiencies, and hereditary hemoglobinopathies. Because the reported association between maternal anemia and preterm labor in different studies is varied depending on the month of pregnancy this study aims to determine this relationship after adjustment for potential confounders. METHODS A case-control study was conducted among 801 pregnant women (267 cases and 534 controls) in Sardashat, Iran from October 2012 to October 2013. Hemoglobin (Hb) values were measured for all women participating in the study during the first and second trimesters of pregnancy, and the average Hb values were presented. Statistical analyzes were performed with logistic regression. RESULTS The mean age of participants was 26.4 ± 5.25 years for the case group and 27.2 ± 6.51 years for the control group. Preterm birth was associated with maternal anemia during pregnancy [aOR = 2.69 (95% CI; 1.46 to 4.95)] even after adjusting for confounding effects including maternal age, history of abortion [aOR = 2.41 (95% CI; 1.42 to 4.08)], history of preterm birth [aOR = 11.38 (95% CI; 3.48 to 37.22)], obesity (aOR: 3.441; CI95%: 1.18-10.06), parity [aOR = 0.42 (95% CI; 0.25 to 0.69)], preeclampsia/eclampsia [aOR = 6.08 (95% CI; 2.64 to 14)], and GDM [aOR = 4.80 (95% CI; 2.02 to 11.41)]). CONCLUSION Early detection and adequate treatment of anemia during pregnancy can help reduce the prevalence of preterm birth.
Collapse
Affiliation(s)
- Rozhan Khezri
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| | - Shaker Salarilak
- Associate Professor of Epidemiology, Department of Public Health, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Sepideh Jahanian
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
23
|
Wu Q. Natriuretic Peptide Signaling in Uterine Biology and Preeclampsia. Int J Mol Sci 2023; 24:12309. [PMID: 37569683 PMCID: PMC10418983 DOI: 10.3390/ijms241512309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Endometrial decidualization is a uterine process essential for spiral artery remodeling, embryo implantation, and trophoblast invasion. Defects in endometrial decidualization and spiral artery remodeling are important contributing factors in preeclampsia, a major disorder in pregnancy. Atrial natriuretic peptide (ANP) is a cardiac hormone that regulates blood volume and pressure. ANP is also generated in non-cardiac tissues, such as the uterus and placenta. In recent human genome-wide association studies, multiple loci with genes involved in natriuretic peptide signaling are associated with gestational hypertension and preeclampsia. In cellular experiments and mouse models, uterine ANP has been shown to stimulate endometrial decidualization, increase TNF-related apoptosis-inducing ligand expression and secretion, and enhance apoptosis in arterial smooth muscle cells and endothelial cells. In placental trophoblasts, ANP stimulates adenosine 5'-monophosphate-activated protein kinase and the mammalian target of rapamycin complex 1 signaling, leading to autophagy inhibition and protein kinase N3 upregulation, thereby increasing trophoblast invasiveness. ANP deficiency impairs endometrial decidualization and spiral artery remodeling, causing a preeclampsia-like phenotype in mice. These findings indicate the importance of natriuretic peptide signaling in pregnancy. This review discusses the role of ANP in uterine biology and potential implications of impaired ANP signaling in preeclampsia.
Collapse
Affiliation(s)
- Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| |
Collapse
|
24
|
Honigberg MC, Truong B, Khan RR, Xiao B, Bhatta L, Vy HMT, Guerrero RF, Schuermans A, Selvaraj MS, Patel AP, Koyama S, Cho SMJ, Vellarikkal SK, Trinder M, Urbut SM, Gray KJ, Brumpton BM, Patil S, Zöllner S, Antopia MC, Saxena R, Nadkarni GN, Do R, Yan Q, Pe'er I, Verma SS, Gupta RM, Haas DM, Martin HC, van Heel DA, Laisk T, Natarajan P. Polygenic prediction of preeclampsia and gestational hypertension. Nat Med 2023; 29:1540-1549. [PMID: 37248299 PMCID: PMC10330886 DOI: 10.1038/s41591-023-02374-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/26/2023] [Indexed: 05/31/2023]
Abstract
Preeclampsia and gestational hypertension are common pregnancy complications associated with adverse maternal and child outcomes. Current tools for prediction, prevention and treatment are limited. Here we tested the association of maternal DNA sequence variants with preeclampsia in 20,064 cases and 703,117 control individuals and with gestational hypertension in 11,027 cases and 412,788 control individuals across discovery and follow-up cohorts using multi-ancestry meta-analysis. Altogether, we identified 18 independent loci associated with preeclampsia/eclampsia and/or gestational hypertension, 12 of which are new (for example, MTHFR-CLCN6, WNT3A, NPR3, PGR and RGL3), including two loci (PLCE1 and FURIN) identified in the multitrait analysis. Identified loci highlight the role of natriuretic peptide signaling, angiogenesis, renal glomerular function, trophoblast development and immune dysregulation. We derived genome-wide polygenic risk scores that predicted preeclampsia/eclampsia and gestational hypertension in external cohorts, independent of clinical risk factors, and reclassified eligibility for low-dose aspirin to prevent preeclampsia. Collectively, these findings provide mechanistic insights into the hypertensive disorders of pregnancy and have the potential to advance pregnancy risk stratification.
Collapse
Affiliation(s)
- Michael C Honigberg
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | - Buu Truong
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Raiyan R Khan
- Department of Computer Science, Columbia University, New York, NY, USA
| | - Brenda Xiao
- University of Pennsylvania, Philadelphia, PA, USA
| | - Laxmi Bhatta
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Ha My T Vy
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rafael F Guerrero
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Art Schuermans
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Faculty of Medicine, KU Leuven, Leuven, Belgium
| | - Margaret Sunitha Selvaraj
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Aniruddh P Patel
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Satoshi Koyama
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - So Mi Jemma Cho
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Integrative Research Center for Cerebrovascular and Cardiovascular Diseases, Seoul, Republic of Korea
| | - Shamsudheen Karuthedath Vellarikkal
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Mark Trinder
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah M Urbut
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Kathryn J Gray
- Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Ben M Brumpton
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU, Norwegian University of Science and Technology, Levanger, Norway
| | - Snehal Patil
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Mariah C Antopia
- Department of Integrative Biology, University of Texas at San Antonio, San Antonio, TX, USA
| | - Richa Saxena
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Girish N Nadkarni
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qi Yan
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | - Itsik Pe'er
- Department of Computer Science, Columbia University, New York, NY, USA
| | | | - Rajat M Gupta
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - David M Haas
- Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hilary C Martin
- Department of Human Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - David A van Heel
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Triin Laisk
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Pradeep Natarajan
- Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
25
|
Nurkkala J, Kauko A, FinnGen, Laivuori H, Saarela T, Tyrmi JS, Vaura F, Cheng S, Bello NA, Aittokallio J, Niiranen T. Associations of polygenic risk scores for preeclampsia and blood pressure with hypertensive disorders of pregnancy. J Hypertens 2023; 41:380-387. [PMID: 36947680 PMCID: PMC9894151 DOI: 10.1097/hjh.0000000000003336] [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: 09/16/2022] [Accepted: 11/01/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Preexisting hypertension increases risk for preeclampsia. We examined whether a generic blood pressure polygenic risk score (BP-PRS), compared with a preeclampsia-specific polygenic risk score (PE-PRS), could better predict hypertensive disorders of pregnancy. METHODS Our study sample included 141 298 genotyped FinnGen study participants with at least one childbirth and followed from 1969 to 2021. We calculated PRSs for SBP and preeclampsia using summary statistics for greater than 1.1 million single nucleotide polymorphisms. RESULTS We observed 8488 cases of gestational hypertension (GHT) and 6643 cases of preeclampsia. BP-PRS was associated with GHT [multivariable-adjusted hazard ratio for 1SD increase in PRS (hazard ratio 1.38; 95% CI 1.35-1.41)] and preeclampsia (1.26, 1.23-1.29), respectively. The PE-PRS was also associated with GHT (1.16; 1.14-1.19) and preeclampsia (1.21, 1.18-1.24), but with statistically more modest magnitudes of effect (P = 0.01). The model c-statistic for preeclampsia improved when PE-PRS was added to clinical risk factors (P = 4.6 × 10-15). Additional increment in the c-statistic was observed when BP-PRS was added to a model already including both clinical risk factors and PE-PRS (P = 1.1 × 10-14). CONCLUSION BP-PRS is strongly associated with hypertensive disorders of pregnancy. Our current observations suggest that the BP-PRS could capture the genetic architecture of preeclampsia better than the current PE-PRSs. These findings also emphasize the common pathways in the development of all BP disorders. The clinical utility of a BP-PRS for preeclampsia prediction warrants further investigation.
Collapse
Affiliation(s)
- Jouko Nurkkala
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital
- Department of Anesthesiology and Intensive Care
| | - Anni Kauko
- Department of Internal Medicine, University of Turku, Turku
| | | | - Hannele Laivuori
- Department of Obstetrics and Gynecology, Tampere University Hospital
- Center for Child, Adolescent, and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University, Tampere
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science, University of Helsinki
- Medical and Clinical Genetics, University of Helsinki and Helsinki University Hospital, Helsinki
| | - Tanja Saarela
- Department of Clinical Genetics, Kuopio University Hospital, Kuopio
| | - Jaakko S. Tyrmi
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Felix Vaura
- Department of Internal Medicine, University of Turku, Turku
| | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Division of Cardiology Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Natalie A. Bello
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jenni Aittokallio
- Division of Perioperative Services, Intensive Care and Pain Medicine, Turku University Hospital
- Department of Anesthesiology and Intensive Care
| | - Teemu Niiranen
- Department of Internal Medicine, University of Turku, Turku
- Division of Medicine, Turku University Hospital
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Turku, Finland
| |
Collapse
|
26
|
Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Beaton AZ, Boehme AK, Buxton AE, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Fugar S, Generoso G, Heard DG, Hiremath S, Ho JE, Kalani R, Kazi DS, Ko D, Levine DA, Liu J, Ma J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Virani SS, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2023 Update: A Report From the American Heart Association. Circulation 2023; 147:e93-e621. [PMID: 36695182 DOI: 10.1161/cir.0000000000001123] [Citation(s) in RCA: 1924] [Impact Index Per Article: 962.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2023 Statistical Update is the product of a full year's worth of effort in 2022 by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. The American Heart Association strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional COVID-19 (coronavirus disease 2019) publications, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
27
|
Woolner AM, Bhattacharya S. Intergenerational trends in reproduction: Infertility and pregnancy loss. Best Pract Res Clin Obstet Gynaecol 2023; 86:102305. [PMID: 36639284 DOI: 10.1016/j.bpobgyn.2022.102305] [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: 08/31/2022] [Revised: 11/25/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022]
Abstract
This review article summarises the evidence for intergenerational trends observed to date within infertility and pregnancy loss. There appears to be evidence of intergenerational trends between mothers and daughters for the age at menopause, endometriosis, polycystic ovarian syndrome (PCOS), male factor infertility and miscarriage. At present, there is no evidence for a predisposition to stillbirth between mothers and daughters. One study found an association with familial predisposition for ectopic pregnancy. Very few studies have considered the potential for paternal transmission of risk of infertility or pregnancy loss. The majority of studies to date have significant limitations because of their observational design, risk of recall bias and risk of confounding. Therefore, high-quality well-designed research, with multi-centre collaboration and utilisation of registry-based data sources and individual patient data, is needed to understand whether infertility and pregnancy loss may have heritable factors. Epidemiological findings need to be followed up and investigated with translational research to determine the possible causalities as well as any implications for clinical practice.
Collapse
Affiliation(s)
- Andrea Mf Woolner
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, United Kingdom.
| | - Siladitya Bhattacharya
- Aberdeen Centre for Women's Health Research, Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Scotland, United Kingdom.
| |
Collapse
|
28
|
Zhao L, Xin S, Wu Y, Huang S, Xu K, Xu Y, Ruan D, Wu B, Chen D, He X. Global DNA and protein interactomes of FLT1P1 (Fms-related tyrosine kinase 1 pseudogene 1) revealed its molecular regulatory functions associated with preeclampsia. Mol Biol Rep 2023; 50:1267-1279. [PMID: 36451001 DOI: 10.1007/s11033-022-08070-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/31/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Preeclampsia (PE) is one of the most serious pregnancy complications with unknown pathogenesis. Emerging evidence has demonstrated that Fms-related tyrosine kinase 1 (FLT1) is highly involved in PE development. As a pseudogene of FLT1, FLT1P1 increased in PE samples. However, its functions remain largely unknown. METHODS AND RESULTS In this study, co-expression analysis was performed to identify the potential target genes of FTL1P1. Then chromatin isolation using RNA purification (ChIRP) method was employed to explore the interactomes of FLT1P1, including interacting with DNA fragments and proteins. We found that in PE samples, both FLT1P1 and FLT1 were highly expressed and closely correlated. ChIRP-protein data revealed that FLT1P1 interacts with translation- and transcription-related proteins, including 4 transcription factors (TFs). ChIRP-DNA analysis revealed that FLT1P1 preferentially interacted with DNA fragments downstream of transcription start sites (TSSs). Functional analysis of its interacting genes revealed that they were enriched in transcriptional regulation and apoptosis-related pathways. Twenty-six TFs, including CREB1 and SRF, were extracted from the potential FLT1P1-interacting gene sets and were potential targets of FLT1P1. CREB1 could bind to FLT1 promoter, and was negatively correlated with FLT1 at the expression level, making it a potential regulator of FLT1. CONCLUSIONS Our study extensively investigated the interactome profiles of FLT1P1, especially the prompter region of TF gene CREB1, and revealed the potential molecular regulatory mechanisms of FLT1 expression in PE samples. Our results provide a novel view of PE pathogenesis, and suggest that FLT1P1 could serve as a potential therapeutic target in PE diagnosis and treatment.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Siming Xin
- Department of Obstetrics, Maternal, Child Health Hospital Afflicted to Nanchang University, Nanchang, People's Republic of China
| | - Yunfei Wu
- Center for Genome Analysis, Wuhan Ruixing Biotechnology Co., Ltd., Wuhan, People's Republic of China
| | - Shaofang Huang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Kangxiang Xu
- Second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Yuqi Xu
- Second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Dong Ruan
- Second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Bingqi Wu
- Second Clinical Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Dong Chen
- Center for Genome Analysis, Wuhan Ruixing Biotechnology Co., Ltd., Wuhan, People's Republic of China
| | - Xiaoju He
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China.
| |
Collapse
|
29
|
Abramova M, Churnosova M, Efremova O, Aristova I, Reshetnikov E, Polonikov A, Churnosov M, Ponomarenko I. Effects of Pre-Pregnancy Overweight/Obesity on the Pattern of Association of Hypertension Susceptibility Genes with Preeclampsia. Life (Basel) 2022; 12:life12122018. [PMID: 36556383 PMCID: PMC9784908 DOI: 10.3390/life12122018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to explore the effects of pre-pregnancy overweight/obesity on the pattern of association of hypertension susceptibility genes with preeclampsia (PE). Ten single-nucleotide polymorphisms (SNPs) of the 10 genome-wide association studies (GWAS)-significant hypertension/blood pressure (BP) candidate genes were genotyped in 950 pregnant women divided into two cohorts according to their pre-pregnancy body mass index (preBMI): preBMI ≥ 25 (162 with PE and 159 control) and preBMI < 25 (290 with PE and 339 control). The PLINK software package was utilized to study the association (analyzed four genetic models using logistic regression). The functionality of PE-correlated loci was analyzed by performing an in silico database analysis. Two SNP hypertension/BP genes, rs805303 BAG6 (OR: 0.36−0.66) and rs167479 RGL3 (OR: 1.86), in subjects with preBMI ≥ 25 were associated with PE. No association between the studied SNPs and PE in the preBMI < 25 group was determined. Further analysis showed that two PE-associated SNPs are functional (have weighty eQTL, sQTL, regulatory, and missense values) and could be potentially implicated in PE development. In conclusion, this study was the first to discover the modifying influence of overweight/obesity on the pattern of association of GWAS-significant hypertension/BP susceptibility genes with PE: these genes are linked with PE in preBMI ≥ 25 pregnant women and are not PE-involved in the preBMI < 25 group.
Collapse
Affiliation(s)
- Maria Abramova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Olesya Efremova
- Department of Medical Genetics, Kharkiv National Medical University, 61022 Kharkov, Ukraine
- Grishchenko Clinic of Reproductive Medicine, 61052 Kharkov, Ukraine
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| | - Alexey Polonikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
- Department of Biology, Medical Genetics and Ecology and Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
- Correspondence:
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia
| |
Collapse
|
30
|
Perišić MM, Vladimir K, Karpov S, Štorga M, Mostashari A, Khanin R. Polygenic Risk Score and Risk Factors for Preeclampsia and Gestational Hypertension. J Pers Med 2022; 12:1826. [PMID: 36579561 PMCID: PMC9694636 DOI: 10.3390/jpm12111826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/23/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Preeclampsia and gestational hypertensive disorders (GHD) are common complications of pregnancy that adversely affect maternal and offspring health, often with long-term consequences. High BMI, advanced age, and pre-existing conditions are known risk factors for GHD. Yet, assessing a woman's risk of GHD based on only these characteristics needs to be reevaluated in order to identify at-risk women, facilitate early diagnosis, and implement lifestyle recommendations. This study demonstrates that a risk score developed with machine learning from the case-control genetics dataset can be used as an early screening test for GHD. We further confirm BMI as a risk factor for GHD and investigate a relationship between GHD and genetically constructed anthropometric measures and biomarkers. Our results show that polygenic risk score can be used as an early screening tool that, together with other known risk factors and medical history, would assist in identifying women at higher risk of GHD before its onset to enable stratification of patients into low-risk and high-risk groups for monitoring and preventative programs to mitigate the risks.
Collapse
Affiliation(s)
- Marija Majda Perišić
- LifeNome Inc., New York, NY 10018, USA
- Faculty of Mechanical Engineering and Naval Architecture, University of Zagreb, 10000 Zagreb, Croatia
| | - Klemo Vladimir
- LifeNome Inc., New York, NY 10018, USA
- Faculty of Electrical Engineering and Computing, University of Zagreb, 10000 Zagreb, Croatia
| | | | - Mario Štorga
- LifeNome Inc., New York, NY 10018, USA
- Faculty of Mechanical Engineering and Naval Architecture, University of Zagreb, 10000 Zagreb, Croatia
| | | | - Raya Khanin
- LifeNome Inc., New York, NY 10018, USA
- Bioinformatics Core, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
31
|
Churnosov M, Abramova M, Reshetnikov E, Lyashenko IV, Efremova O, Churnosova M, Ponomarenko I. Polymorphisms of hypertension susceptibility genes as a risk factors of preeclampsia in the Caucasian population of central Russia. Placenta 2022; 129:51-61. [PMID: 36219912 DOI: 10.1016/j.placenta.2022.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/18/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The study was designed to assess the effects of hypertension (HT) susceptibility genes polymorphisms in the development of preeclampsia (PE) in Caucasians from Central Russia. METHODS PE patients (n = 452) and women control group (n = 498) were genotyped for 10 polymorphisms of HT/blood pressure (BP) susceptibility genes (according to the previously published GWAS in Caucasian populations) including AC026703.1 (rs1173771), HFE (rs1799945), BAG6 (rs805303), PLCE1 (rs932764), OBFC1 (rs4387287), ARHGAP42 (rs633185), CERS5 (rs7302981), ATP2B1 (rs2681472), TBX2 (rs8068318) and RGL3 (rs167479). A logistic regression method was applied to search for associations between SNPs and PE. The relationship between SNP-SNP interactions and PE risk was analyzed by performing MB-MDR. RESULTS The rs1799945 gene in HFE significantly independently increased the risk of developing PE (OR = 2.24) and rs805303 in BAG6 was associated with a reduced risk in the occurrence of PE (OR = 0.55-0.78). Among the 10 SNPs examined, nine SNPs were associated with PEs within the 10 most significant SNP-SNP interaction models. Loci rs7302981 CERS5, rs805303 BAG6 and rs932764 PLCE1 contributed to the largest number of epistatic models (50% or more). DISCUSSION The present study is the first to report an association between polymorphisms of HT/BP susceptibility genes important for GWAS and the risk of PE in Caucasians from Central Russia. Our pathway-based functional annotation of the PE risk variants highlights the potential regulatory function (epigenetic/eQTL/sQTL/non-synonymous) that nine genetic risk markers and their 115 highly correlated variants exert on 155 genes. The study shows that these genes may function cooperatively in key signaling pathways in PE biology.
Collapse
Affiliation(s)
- Mikhail Churnosov
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia.
| | - Maria Abramova
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia
| | - Evgeny Reshetnikov
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia
| | - Igor V Lyashenko
- Belgorod State National Research University, Department of English Philology and Cross-cultural Communication, Belgorod, Russia
| | - Olesya Efremova
- Kharkiv National Medical University, Department of Medical Genetics, Kharkov, Ukraine; Grishchenko Clinic of Reproductive Medicine, Kharkov, Ukraine
| | - Maria Churnosova
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia
| | - Irina Ponomarenko
- Belgorod State National Research University, Department of Medical Biological Disciplines, Belgorod, Russia
| |
Collapse
|
32
|
Moffett A, Shreeve N. Local immune recognition of trophoblast in early human pregnancy: controversies and questions. Nat Rev Immunol 2022; 23:222-235. [PMID: 36192648 PMCID: PMC9527719 DOI: 10.1038/s41577-022-00777-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 02/02/2023]
Abstract
The role of the maternal immune system in reproductive success in humans remains controversial. Here we focus on the events that occur in the maternal decidua during the first few weeks of human pregnancy, because this is the site at which maternal leukocytes initially interact with and can recognize fetal trophoblast cells, potentially involving allorecognition by both T cells and natural killer (NK) cells. NK cells are the dominant leukocyte population in first-trimester decidua, and genetic studies point to a role of allorecognition by uterine NK cells in establishing a boundary between the mother and the fetus. By contrast, definitive evidence that allorecognition by decidual T cells occurs during the first trimester is lacking. Thus, our view is that during the crucial period when the placenta is established, damaging T cell-mediated adaptive immune responses towards placental trophoblast are minimized, whereas NK cell allorecognition contributes to successful implantation and healthy pregnancy.
Collapse
Affiliation(s)
- Ashley Moffett
- grid.5335.00000000121885934Department of Pathology, University of Cambridge, Cambridge, UK
| | - Norman Shreeve
- grid.5335.00000000121885934Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
| |
Collapse
|
33
|
Zhang M, Deng X, Jiang Z, Ge Z. Identification of underlying mechanisms and hub gene-miRNA networks of the genomic subgroups in preeclampsia development. Medicine (Baltimore) 2022; 101:e29569. [PMID: 35866827 PMCID: PMC9302342 DOI: 10.1097/md.0000000000029569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Preeclampsia is a hypertensive disorder of pregnancy that can lead to multiorgan complications in the mother and fetus. Our study aims to uncover the underlying mechanisms and hub genes between genomic subgroups of preeclampsia. A total of 180 preeclampsia cases from 4 gene profiles were classified into 3 subgroups. Weighted gene coexpression analysis was performed to uncover the genomic characteristics associated with different clinical features. Functional annotation was executed within the significant modules and hub genes were predicted using Cytoscape software. Subsequently, miRNet analysis was performed to identify potential miRNA-mRNA networks. Three key subgroup-specific modules were identified. Patients in subgroup II were found to develop more severe preeclampsia symptoms. Subgroup II, characterized by classical markers, was considered representative of typical preeclampsia patients. Subgroup I was considered as an early stage of preeclampsia with normal-like gene expression patterns. Moreover, subgroup III was a proinflammatory subgroup, which presented immune-related genomic characteristics. Subsequently, miR-34a-5p and miR-106a-5p were found to be correlated with all 3 significant gene modules. This study revealed the transcriptome classification of preeclampsia cases with unique gene expression patterns. Potential hub genes and miRNAs may facilitate the identification of therapeutic targets for preeclampsia in future.
Collapse
Affiliation(s)
- Min Zhang
- Department of Obstetrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaheng Deng
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ziyan Jiang
- Department of Obstetrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhiping Ge
- Department of Obstetrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Zhiping Ge, Department of Obstetrics, The First Affiliated Hospital of Nanjing Medical University, 368 Jiangdong North Road, Nanjing 210000, China. (e-mail: )
| |
Collapse
|
34
|
Cai X, Liang N, Cai X, Zhou Q, Dang Q, Hu Z, Yu H. Lipid Metabolic Genes and Maternal Supraphysiological Hypercholesterolemia: An Analysis of Maternal-fetal Interaction. J Clin Endocrinol Metab 2022; 107:e3134-e3144. [PMID: 35575245 DOI: 10.1210/clinem/dgac317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Indexed: 11/19/2022]
Abstract
CONTEXT The joint associations of maternal and fetal single nucleotide polymorphisms (SNPs) of lipid metabolic genes with the risk of maternal supraphysiological hypercholesterolemia (MSPH) are unclear. OBJECTIVE This study aims to investigate the associations of maternal/fetal SNPs of APOE, LPL, LDLR, PCSK9, and SCARB1 with the risk of MSPH and explore whether the maternal-fetal pairing pattern of the risk alleles can affect MSPH risk. METHODS A nested case-control study was conducted that included 182 pregnant women with MSPH and 182 with maternal physiological hypercholesterolemia. Maternal venous and umbilical venous blood were collected to detect the SNPs of genes. The primary outcome was MSPH. Logistic regression model was used to determine the associations of SNPs with risk of MSPH. RESULTS The C-allele in maternal APOE rs429358 T > C (adjusted odds ratio [OR] = 1.72, P = 0.033), G-allele in fetal APOE rs440446 C > G (adjusted OR = 1.62, P = 0.012) and T-allele in fetal LPL rs263 C > T (adjusted OR = 1.53, P = 0.011) increased the risk of MSPH. The A-allele in maternal LDLR rs7258950 G > A decreased the risk of MSPH (adjusted OR = 0.67, P = 0.028). For maternal-fetal pairing analysis, the variant concordance of PCSK9 rs2149041, rs7523141, rs7523242, rs7525649, and LDLR rs7258950 were associated with the decreased risk of MSPH under the dominant model. The variant concordance of other SNPs of PCSK9, APOE, LDLR, LPL, and SCARB1 were associated with the increased risk of MSPH. CONCLUSION This study supports the hypothesis that maternal and fetal genetic polymorphisms of lipid metabolic genes are associated with the risk of MSPH. The maternal-fetal variant concordance is also associated with this risk.
Collapse
Affiliation(s)
- Xiaxia Cai
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Ning Liang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
- Department of Clinical Nutrition, Haidian Maternal and Child Health Hospital, Beijing 100080, P. R. China
| | - Xueping Cai
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
- Department of Clinical Nutrition, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361004, P. R. China
| | - Qi Zhou
- Department of Gynaecology and Obstetrics, Xuanwu Hospital of Capital Medical University, Beijing 100053, P. R. China
| | - Qinyu Dang
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Zhuo Hu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| | - Huanling Yu
- School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P. R. China
| |
Collapse
|
35
|
Abstract
Hypertensive disorders of pregnancy, including gestational hypertension, preeclampsia, and eclampsia, are a worldwide health problem. Hypertensive disorders of pregnancy affect more than 10% of pregnancies and are associated with increased mortality and morbidity for both mother and fetus. Although patients' outcomes and family's experience will always be the primary concern regarding hypertensive complications during pregnancy, the economic aspect of this disease is also worth noting. Compared with normotensive pregnancies, those related with hypertension resulted in an excess increase in hospitalization and healthcare cost. Hence, the focus of this review is to analyze hypertensive disorders of pregnancy and to present practical tips with clear instructions for the clinical management of hypertensive disorders of pregnancy. This overview offers a detailed approach from the diagnosis to treatment and follow-up of a pregnant women with hypertension, evidence based, to support these instructions.
Collapse
|
36
|
Chen Z, Gan J, Zhang M, Du Y, Zhao H. Ferroptosis and Its Emerging Role in Pre-Eclampsia. Antioxidants (Basel) 2022; 11:1282. [PMID: 35883776 PMCID: PMC9312356 DOI: 10.3390/antiox11071282] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 11/27/2022] Open
Abstract
Iron is essential for cell survival, and iron deficiency is a known risk factor for many reproductive diseases. Paradoxically, such disorders are also more common in cases of iron overload. Here, we evaluated the role of ferroptosis in women's health, particularly focusing on pre-eclampsia (PE). PE is a multisystem disorder and is one of the leading causes of maternal and perinatal morbidity and mortality, especially when the condition is of early onset. Nevertheless, the exact etiological mechanism of PE remains unclear. Interestingly, ferroptosis, as a regulated iron-dependent cell death pathway, involves a lethal accumulation of lipid peroxides and shares some characteristics with PE pathophysiology. In this review, we comprehensively reviewed and summarized recent studies investigating the molecular mechanisms involved in the regulation and execution of ferroptosis, as well as ferroptosis mechanisms in the pathology of PE. We propose that ferroptosis not only plays an important role in PE, but may also become a novel therapeutic target for PE.
Collapse
Affiliation(s)
- Zhixian Chen
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (Z.C.); (J.G.); (M.Z.)
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Jianfeng Gan
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (Z.C.); (J.G.); (M.Z.)
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Mo Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (Z.C.); (J.G.); (M.Z.)
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Yan Du
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (Z.C.); (J.G.); (M.Z.)
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| | - Hongbo Zhao
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China; (Z.C.); (J.G.); (M.Z.)
- Department of Obstetrics and Gynecology of Shanghai Medical School, Fudan University, Shanghai 200032, China
| |
Collapse
|
37
|
Nieves-Colón MA, Badillo Rivera KM, Sandoval K, Villanueva Dávalos V, Enriquez Lencinas LE, Mendoza-Revilla J, Adhikari K, González-Buenfil R, Chen JW, Zhang ET, Sockell A, Ortiz-Tello P, Hurtado GM, Condori Salas R, Cebrecos R, Manzaneda Choque JC, Manzaneda Choque FP, Yábar Pilco GP, Rawls E, Eng C, Huntsman S, Burchard E, Ruiz-Linares A, González-José R, Bedoya G, Rothhammer F, Bortolini MC, Poletti G, Gallo C, Bustamante CD, Baker JC, Gignoux CR, Wojcik GL, Moreno-Estrada A. Clotting factor genes are associated with preeclampsia in high-altitude pregnant women in the Peruvian Andes. Am J Hum Genet 2022; 109:1117-1139. [PMID: 35588731 PMCID: PMC9247825 DOI: 10.1016/j.ajhg.2022.04.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Preeclampsia is a multi-organ complication of pregnancy characterized by sudden hypertension and proteinuria that is among the leading causes of preterm delivery and maternal morbidity and mortality worldwide. The heterogeneity of preeclampsia poses a challenge for understanding its etiology and molecular basis. Intriguingly, risk for the condition increases in high-altitude regions such as the Peruvian Andes. To investigate the genetic basis of preeclampsia in a population living at high altitude, we characterized genome-wide variation in a cohort of preeclamptic and healthy Andean families (n = 883) from Puno, Peru, a city located above 3,800 meters of altitude. Our study collected genomic DNA and medical records from case-control trios and duos in local hospital settings. We generated genotype data for 439,314 SNPs, determined global ancestry patterns, and mapped associations between genetic variants and preeclampsia phenotypes. A transmission disequilibrium test (TDT) revealed variants near genes of biological importance for placental and blood vessel function. The top candidate region was found on chromosome 13 of the fetal genome and contains clotting factor genes PROZ, F7, and F10. These findings provide supporting evidence that common genetic variants within coagulation genes play an important role in preeclampsia. A selection scan revealed a potential adaptive signal around the ADAM12 locus on chromosome 10, implicated in pregnancy disorders. Our discovery of an association in a functional pathway relevant to pregnancy physiology in an understudied population of Native American origin demonstrates the increased power of family-based study design and underscores the importance of conducting genetic research in diverse populations.
Collapse
Affiliation(s)
- Maria A Nieves-Colón
- Laboratorio Nacional de Genómica para la Biodiversidad (UGA-LANGEBIO), CINVESTAV, Irapuato, Guanajuato 36821, México; School of Human Evolution and Social Change, Arizona State University, Tempe, AZ 85281, USA; Department of Anthropology, University of Minnesota Twin Cities, Minneapolis, MN 55455, USA.
| | | | - Karla Sandoval
- Laboratorio Nacional de Genómica para la Biodiversidad (UGA-LANGEBIO), CINVESTAV, Irapuato, Guanajuato 36821, México
| | | | | | - Javier Mendoza-Revilla
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru; Human Evolutionary Genetics Unit, Institut Pasteur, UMR 2000, CNRS, Paris 75015, France
| | - Kaustubh Adhikari
- School of Mathematics and Statistics, Faculty of Science, Technology, Engineering and Mathematics, The Open University, Milton Keynes MK7 6AA, UK; Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, WC1E 6BT London, UK
| | - Ram González-Buenfil
- Laboratorio Nacional de Genómica para la Biodiversidad (UGA-LANGEBIO), CINVESTAV, Irapuato, Guanajuato 36821, México
| | - Jessica W Chen
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Elisa T Zhang
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Alexandra Sockell
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA
| | | | - Gloria Malena Hurtado
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Ramiro Condori Salas
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Ricardo Cebrecos
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | | | | | | | - Erin Rawls
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ 85281, USA
| | - Celeste Eng
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Scott Huntsman
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Esteban Burchard
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Andrés Ruiz-Linares
- Department of Genetics, Evolution and Environment, and UCL Genetics Institute, University College London, WC1E 6BT London, UK; Aix-Marseille Université, CNRS, EFS, ADES, 13005 Marseille, France; Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Yangpu District, Shanghai, China
| | - Rolando González-José
- Instituto Patagónico de Ciencias Sociales y Humanas, Centro Nacional Patagónico-CONICET y Programa Nacional de Referencia y Biobanco Genómico de la Población Argentina (PoblAr), Ministerio de Ciencia, Tecnología e Innovación, Puerto Madryn, Chubut, Argentina
| | - Gabriel Bedoya
- Genética Molecular (GENMOL), Universidad de Antioquía, Medellin, Colombia
| | - Francisco Rothhammer
- Instituto de Alta Investigación Universidad de Tarapacá, Tarapacá, Chile; Programa de Genética Humana, ICBM Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Maria Cátira Bortolini
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Caixa Postal 15053, 91501-970 Porto Alegre, Rio Grande do Sul, Brazil
| | - Giovanni Poletti
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Carla Gallo
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Carlos D Bustamante
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Department of Biomedical Data Science, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Julie C Baker
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA
| | | | - Genevieve L Wojcik
- Department of Epidemiology, Bloomberg School of Public Health, John Hopkins University, Baltimore, MD 21205, USA
| | - Andrés Moreno-Estrada
- Laboratorio Nacional de Genómica para la Biodiversidad (UGA-LANGEBIO), CINVESTAV, Irapuato, Guanajuato 36821, México.
| |
Collapse
|
38
|
Schuster J, Tollefson GA, Zarate V, Agudelo A, Stabila J, Ragavendran A, Padbury J, Uzun A. Protein Network Analysis of Whole Exome Sequencing of Severe Preeclampsia. Front Genet 2022; 12:765985. [PMID: 35719905 PMCID: PMC9201216 DOI: 10.3389/fgene.2021.765985] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/03/2021] [Indexed: 11/13/2022] Open
Abstract
Preeclampsia is a hypertensive disorder of pregnancy, which complicates up to 15% of US deliveries. It is an idiopathic disorder associated with several different phenotypes. We sought to determine if the genetic architecture of preeclampsia can be described by clusters of patients with variants in genes in shared protein interaction networks. We performed a case-control study using whole exome sequencing on early onset preeclamptic mothers with severe clinical features and control mothers with uncomplicated pregnancies between 2016 and 2020. A total of 143 patients were enrolled, 61 women with early onset preeclampsia with severe features based on ACOG criteria, and 82 control women at term, matched for race and ethnicity. A network analysis and visualization tool, Proteinarium, was used to confirm there are clusters of patients with shared gene networks associated with severe preeclampsia. The majority of the sequenced patients appear in two significant clusters. We identified one case dominant and one control dominant cluster. Thirteen genes were unique to the case dominated cluster. Among these genes, LAMB2, PTK2, RAC1, QSOX1, FN1, and VCAM1 have known associations with the pathogenic mechanisms of preeclampsia. Using bioinformatic analysis, we were able to identify subsets of patients with shared protein interaction networks, thus confirming our hypothesis about the genetic architecture of preeclampsia.
Collapse
Affiliation(s)
- Jessica Schuster
- Pediatrics, Women and Infants Hospital, Providence, RI, United States
- Pediatrics, Warren Alpert Medical School, Brown University, Providence, RI, United States
| | | | - Valeria Zarate
- Pediatrics, Women and Infants Hospital, Providence, RI, United States
| | - Anthony Agudelo
- Pediatrics, Women and Infants Hospital, Providence, RI, United States
| | - Joan Stabila
- Pediatrics, Women and Infants Hospital, Providence, RI, United States
| | - Ashok Ragavendran
- Center for Computation and Visualization, Brown University, Providence, RI, United States
- Computational Biology of Human Disease, Brown University, Providence, RI, United States
| | - James Padbury
- Pediatrics, Women and Infants Hospital, Providence, RI, United States
- Pediatrics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Center for Computational Molecular Biology, Brown University, Providence, RI, United States
| | - Alper Uzun
- Pediatrics, Women and Infants Hospital, Providence, RI, United States
- Pediatrics, Warren Alpert Medical School, Brown University, Providence, RI, United States
- Computational Biology of Human Disease, Brown University, Providence, RI, United States
- Center for Computational Molecular Biology, Brown University, Providence, RI, United States
| |
Collapse
|
39
|
Alexandrova M, Manchorova D, Dimova T. Immunity at maternal-fetal interface: KIR/HLA (Allo)recognition. Immunol Rev 2022; 308:55-76. [PMID: 35610960 DOI: 10.1111/imr.13087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/28/2022] [Accepted: 05/09/2022] [Indexed: 12/15/2022]
Abstract
Both KIR and HLA are the most variable gene families in the human genome. The recognition of the semi-allogeneic embryo-derived trophoblasts by maternal decidual NK (dNK) cells is essential for the establishment of the functional placenta. This recognition is based on the KIR-HLA interactions and trophoblast expresses a specific HLA profile that constitutes classical polymorphic HLA-C and non-classical oligomorphic HLA-E, HLA-F, and HLA-G molecules. This review highlights some features of the KIR/HLA-C (allo)recognition by decidual NK (dNK) cells as a main immune cell population specifically enriched at maternal-fetal interface during human early pregnancy. How KIR/HLA-C axis operates in pregnancy disorders and in the context of transplacental infections is discussed as well. We summarized old and new data on dNK-cell functional plasticity, their selective expression of KIR and fetal maternal/paternal HLA-C haplotypes present. Results showed that KIR-HLA-C combinations and the corresponding axis operate differently in each pregnancy, determined by the variability of both maternal KIR haplotypes and fetus' maternal/paternal HLA-C allotype combinations. Moreover, the maturation of NK cells strongly depends on if or not HLA allotypes for certain KIR are present. We suggest that the unique KIR/HLA combinations reached in each pregnancy (normal and pathological) should be studied according to well-defined guidelines and unified methodologies to have comparable results ease to interpret and use in clinics.
Collapse
Affiliation(s)
- Marina Alexandrova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Diana Manchorova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Tanya Dimova
- Institute of Biology and Immunology of Reproduction, Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
40
|
Zhang H, Wang W. Risk factors and adverse pregnancy outcomes in older pregnant women with hypertensive disorders of pregnancy. J Obstet Gynaecol Res 2022; 48:1710-1720. [PMID: 35596590 DOI: 10.1111/jog.15295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 04/02/2022] [Accepted: 05/06/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND To assess the independent risk factors and pregnancy outcomes of hypertensive disorders of pregnancy (HDPs) in Chinese advanced age pregnancy women. DESIGN Secondary analysis of international published data. MATERIALS AND METHODS PubMed, Web of Science, Scopus, Embase, Cochrane library, and Chinese databases from inception to February 23, 2022 were searched to identify eligible studies. Meta-analysis was conducted to provide meaningful summative outcomes. The quality evaluation of included study was performed. The analyses were done with Review Manager 5.3 and Stata 14.0 software. RESULTS Totally, 13 eligible articles were included, among which 3 articles investigated the risk factors, 6 articles studied pregnancy outcomes, and 4 articles examined both risk factors and pregnancy outcomes. Chinese older pregnant women with the HDPs had high pooled incidences of abnormal body mass index, maternal education, family history of hypertension, history of chronic hypertension, gestational diabetes, irregular delivery inspection, psychosocial factors, and primiparity compared to those without HDPs. The incidence of cesarean delivery, placental abruption, premature rupture of membranes, postpartum hemorrhage, preterm birth, fetal asphyxia, fetal distress, fetal growth restriction, and low birth weight in Chinese older pregnant women with HPDs were significantly higher than those without HDPs. CONCLUSION This study identified several possible risk factors which promoted the onset of HDPs in Chinese older pregnant women, and HDPs significantly increases risk of adverse pregnancy outcomes (APOs). Further research is needed to take targeted measures to prevent HDPs and explore efficient management methods to minimize the risk of APOs associated with HDPs in Chinese older pregnant women.
Collapse
Affiliation(s)
- Huijuan Zhang
- International Inpatient Department, Zhongshan Hospital Affiliated to Dalian University, Dalian City, Liaoning Province, China
| | - Wei Wang
- International Inpatient Department, Zhongshan Hospital Affiliated to Dalian University, Dalian City, Liaoning Province, China
| |
Collapse
|
41
|
Raia-Barjat T, Edebiri O, Ni Ainle F. Preeclampsia and Venous Thromboembolism: Pathophysiology and Potential Therapy. Front Cardiovasc Med 2022; 9:856923. [PMID: 35345482 PMCID: PMC8957069 DOI: 10.3389/fcvm.2022.856923] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 02/11/2022] [Indexed: 12/23/2022] Open
Abstract
Preeclampsia (PET) is a multisystem inflammatory disorder that represents a leading cause of feto-maternal morbidity and mortality, complicating 2-5% of all pregnancies. PET incurs an increased risk of venous thromboembolism, which is one of the leading causes of death in pregnancy and in the postpartum period. This prothrombotic phenotype is attributable to the maternal phase of PET, which is characterized by a systemic inflammatory response and coagulation activation. Research continues to be undertaken in terms of preventative measures, however, currently revolves around pharmacological low dose aspirin initiated in the first trimester of pregnancy for those with risk factors. Treatment involves antenatal corticosteroids for fetal lung development in preterm birth, parenteral magnesium sulfate for fetal neuroprotection and maternal seizure prophylaxis, and timely birth of the fetus and placenta being the only definitive treatment of PET. Patients with a venous thromboembolism (VTE) risk deemed to be >1-3% are treated with pharmacological thromboprophylaxis in the form of low molecular weight heparin. Completing each woman's VTE risk assessment is crucial, particularly in the setting of PET, as there is also a proven associated competing hemorrhagic risk.
Collapse
Affiliation(s)
- Tiphaine Raia-Barjat
- Department of Gynecology and Obstetrics, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Étienne, France
- INSERM U1059, SAINBIOSE, Université Jean Monnet, Saint-Étienne, France
| | - Osasere Edebiri
- Department of Haematology, Mater Misericordiae University Hospital, Rotunda Hospital, Dublin, Ireland
| | - Fionnuala Ni Ainle
- Department of Haematology, Mater Misericordiae University Hospital, Rotunda Hospital, Dublin, Ireland
- UCD School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
42
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 2923] [Impact Index Per Article: 974.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
43
|
Vaura F, Palmu J, Aittokallio J, Kauko A, Niiranen T. Genetic, Molecular, and Cellular Determinants of Sex-Specific Cardiovascular Traits. Circ Res 2022; 130:611-631. [PMID: 35175841 DOI: 10.1161/circresaha.121.319891] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite the well-known sex dimorphism in cardiovascular disease traits, the exact genetic, molecular, and cellular underpinnings of these differences are not well understood. A growing body of evidence currently points at the links between cardiovascular disease traits and the genome, epigenome, transcriptome, and metabolome. However, the sex-specific differences in these links remain largely unstudied due to challenges in bioinformatic methods, inadequate statistical power, analytic costs, and paucity of valid experimental models. This review article provides an overview of the literature on sex differences in genetic architecture, heritability, epigenetic changes, transcriptomic signatures, and metabolomic profiles in relation to cardiovascular disease traits. We also review the literature on the associations between sex hormones and cardiovascular disease traits and discuss the potential mechanisms underlying these associations, focusing on human studies.
Collapse
Affiliation(s)
- Felix Vaura
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Joonatan Palmu
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Jenni Aittokallio
- Department of Anesthesiology and Intensive Care (J.A.), University of Turku, Finland.,Division of Perioperative Services, Intensive Care and Pain Medicine (J.A.), Turku University Hospital, Finland
| | - Anni Kauko
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland
| | - Teemu Niiranen
- Department of Internal Medicine (F.V., J.P., A.K., T.N.), University of Turku, Finland.,Division of Medicine (T.N.), Turku University Hospital, Finland.,Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland (T.N.)
| |
Collapse
|
44
|
Preeclampsia, Natural History, Genes, and miRNAs Associated with the Syndrome. J Pregnancy 2022; 2022:3851225. [PMID: 35198246 PMCID: PMC8860533 DOI: 10.1155/2022/3851225] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/19/2022] [Indexed: 12/13/2022] Open
Abstract
Preeclampsia (PE) is a hypertensive disease that affects pregnant women after 20 weeks of gestation. This disease is associated with an important risk of maternal and fetal mortality. PE is described as a placental pathology because, after delivery, most women recover normal arterial pressure. Poor invasion of the spiral arteries is a phenomenon well described in PE; this leads to a hypoxic uterine bed and imbalance of antiangiogenic and proangiogenic factors in the uteroplacental region, which in turn triggers the disease phenotype. The causes of the pathology are unclear; nevertheless, numerous approaches, including next-generation sequencing, association, and case control and miRNA studies, have shed light on the genetic/molecular basis of PE. These studies help us better understand the disease to advance new treatment strategies.
Collapse
|
45
|
Pruszkowska-Przybylska P, Brennecke S, Moses EK, Melton PE. Evaluation of epigenetic age calculators between preeclampsia and normotensive pregnancies in an Australian cohort. Sci Rep 2022; 12:1664. [PMID: 35102228 PMCID: PMC8803933 DOI: 10.1038/s41598-022-05744-4] [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: 09/11/2021] [Accepted: 01/11/2022] [Indexed: 12/19/2022] Open
Abstract
Advanced biological aging, as assessed through DNA methylation markers, is associated with several complex diseases. The associations between maternal DNA methylation age and preeclampsia (PE) have not been fully assessed. The aim of this study was to examine if increased maternal DNA methylation age (DNAmAge) was shown to be accelerated in women with PE when compared to women who had normotensive pregnancies. The case/control cohort available for study consisted of 166 women (89 with normotensive pregnancy, 77 with PE) recruited previously at the Royal Women’s Hospital in Melbourne, Australia. DNA methylation profiles were obtained using the Illumina EPIC Infinium array for analysis of genomic DNA isolated from whole blood. These profiles were used to calculate seven estimates of DNAmAge and included (1) Horvath, (2) Hannum, (3) Horvath Skin and Blood, (4) Wu, (5) PhenoAge, (6) telomere length and (7) GrimAge and its surrogate measures. Three measures of DNA methylation age acceleration were calculated for all seven measures using linear regression. Pearson's correlation was performed to investigate associations between chronological age and DNAmAge. Differences between chronological age and DNAmAge and epigenetic age acceleration were investigated using t-tests. No significant difference was observed for chronological age between women with PE (age = 30.53 ± 5.68) and women who had normotensive pregnancies (age = 31.76 ± 4.76). All seven DNAmAge measures were significantly correlated (p < 0.001) with chronological age. After accounting for multiple testing and investigating differences in DNAmAge between normotensive women and women with PE, only Wu DNAmAge was significant (p = 0.001). When examining differences for epigenetic age acceleration between PE and normotensive women Hannum, Wu, and PhenoAge DNAmAge estimates (p < 0.001) were significant for both epigenetic age acceleration and intrinsic acceleration models. We found that accelerated maternal DNAmAge is increased in women with PE in some models of epigenetic aging. This research underlines the importance for further investigation into the potential changes of differential DNA methylation in PE.
Collapse
Affiliation(s)
| | - Shaun Brennecke
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, The Royal Women's Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - Eric K Moses
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.,School of Global Population Health, University of Western Australia, Perth, Australia
| | - Phillip E Melton
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.,School of Biomedical Science, University of Western Australia, Perth, Australia
| |
Collapse
|
46
|
Wei X, Yuan Y, Yang Q. SNHG22 promotes migration and invasion of trophoblasts via miR-128-3p/PCDH11X axis and activates PI3K/Akt signaling pathway. Clinics (Sao Paulo) 2022; 77:100055. [PMID: 35679761 PMCID: PMC9188964 DOI: 10.1016/j.clinsp.2022.100055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/21/2022] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Long non-coding RNAs (LncRNAs) act as an indispensable role in the Preeclampsia (PE)-related trophoblast function, while its relationship with Small Nucleolar RNA Host Gene 22 (SNHG22) remains unknown. Hence, this study aimed to investigate the roles of lncRNA SNHG22 in the Preeclampsia (PE)-related trophoblasts function and the underlying mechanism. METHOD Normal placentas and placentas from PE patients were collected to detect the expression of lncRNA SNHG22. Then, trophoblasts HTR-8/Svneo and JEG-3 were purchased, cultured, and treated to investigate the roles of lncRNA SNHG22 on cell migration and invasion as well as its underlying regulatory mechanism. RESULTS The SNHG22 was downregulated in PE patients, and it was found that SNHG22 overexpression could drive migration and invasion of trophoblasts, while SNHG22 depletion exerted a suppressive effect. Mechanistically, SNHG22 was validated to regulate microRNA-128-3p (miR-128-3p), and Protocadherin 11 X-Linked (PCDH11X) was identified as the target gene of miR-128-3p. Furthermore, it was found that SNHG22 acted as a promoter in the migration and invasion of trophoblast cells in a miR-128-3p/PCDH11X dependent manner, and SNHG22 silencing weakened the activation of PCDH11X-mediated PI3K/Akt signaling pathways through inhibiting miR-128-3p, thereby preventing migration and invasion of trophoblasts. CONCLUSION SNHG22 acted as a driver in the migration and invasion of trophoblasts and may be considered a candidate for the amelioration of PE.
Collapse
Affiliation(s)
- Xiaoying Wei
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, China
| | - Yichong Yuan
- Department of Gynaecology, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, China
| | - Qiong Yang
- Department of Obstetrics, Maternal and Child Health Hospital of Hubei Province, Wuhan, Hubei, China.
| |
Collapse
|
47
|
Miller EC, Wilczek A, Bello NA, Tom S, Wapner R, Suh Y. Pregnancy, preeclampsia and maternal aging: From epidemiology to functional genomics. Ageing Res Rev 2022; 73:101535. [PMID: 34871806 PMCID: PMC8827396 DOI: 10.1016/j.arr.2021.101535] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 11/15/2021] [Accepted: 12/01/2021] [Indexed: 01/03/2023]
Abstract
Women live longer than men but experience greater disability and a longer period of illness as they age. Despite clear sex differences in aging, the impact of pregnancy and its complications, such as preeclampsia, on aging is an underexplored area of geroscience. This review summarizes our current knowledge about the complex links between pregnancy and age-related diseases, including evidence from epidemiology, clinical research, and genetics. We discuss the relationship between normal and pathological pregnancy and maternal aging, using preeclampsia as a primary example. We review the results of human genetics studies of preeclampsia, including genome wide association studies (GWAS), and attempted to catalog genes involved in preeclampsia as a gateway to mechanisms underlying an increased risk of later life cardio- and neuro- vascular events. Lastly, we discuss challenges in interpreting the GWAS of preeclampsia and provide a functional genomics framework for future research needed to fully realize the promise of GWAS in identifying targets for geroprotective prevention and therapeutics against preeclampsia.
Collapse
Affiliation(s)
- Eliza C. Miller
- Department of Neurology, Division of Stroke and Cerebrovascular Disease, Columbia University Irving Medical Center, New York, NY, USA
| | - Ashley Wilczek
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA
| | - Natalie A. Bello
- Department of Medicine, Division of Cardiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah Tom
- Department of Neurology, Division of Neurology Clinical Outcomes Research and Population Science and the Department of Epidemiology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ronald Wapner
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA.
| | - Yousin Suh
- Department of Obstetrics and Gynecology, Columbia University Irving Medical Center, New York, NY, USA; Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
48
|
The Impact of PTPRK and ROS1 Polymorphisms on the Preeclampsia Risk in Han Chinese Women. Int J Hypertens 2021; 2021:3275081. [PMID: 34646579 PMCID: PMC8505056 DOI: 10.1155/2021/3275081] [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: 05/07/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023] Open
Abstract
Objective Preeclampsia (PE) is a severe complication in pregnancy and a leading cause of maternal and infant mortality. However, the exact underlying etiology of PE remains unknown. Emerging evidence indicates that the cause of PE is associated with genetic factors. Therefore, the aim of this study is to identify susceptibility genes to PE. Materials and Methods Human Exome BeadChip assays were conducted using 370 cases and 482 controls and 21 loci were discovered. A further independent set of 958 cases and 1007 controls were recruited for genotyping to determine whether the genes of interest ROS1 and PTPRK are associated with PE. Immunohistochemistry was used for localization. Both qPCR and Western blotting were utilized to investigate the levels of PTPRK in placentas of 20 PE and 20 normal pregnancies. Results The allele frequency of PTPRK rs3190930 differed significantly between PE and controls and was particularly significant in severe PE subgroup and early-onset PE subgroup. PTPRK is primarily localized in placental trophoblast cells. The mRNA and protein levels of PTPRK in PE were significantly higher than those in controls. Conclusion These results suggest that PTPRK appears to be a previously unrecognized susceptibility gene for PE in Han Chinese women, and its expression is also associated with PE, while ROS1 rs9489124 has no apparent correlation with PE risk.
Collapse
|
49
|
Guan C, Zhao F, Yang Z, Tang Q, Wang L, Li X, Zhang L, Deng Z, Hou H, Wang J, Xu Y, Zhang R, Lin Y, Tan P, Zhang Y, Liu S, Zhang L. A review of key cytokines based on gene polymorphism in the pathogenesis of pre-eclampsia. Am J Reprod Immunol 2021; 87:e13503. [PMID: 34599631 DOI: 10.1111/aji.13503] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Although a number of theories have been suggested, including roles for oxidative stress, an abnormal maternal-fetal interface, and genetic and environmental factors, the etiopathology of pre-eclampsia (PE) remains unclear. Maternal immune tolerance is important for maintaining pregnancy, and researchers have increasingly focused on the critical roles of cytokines in the pathogenesis of PE in recent years. The assessment of candidate genetic polymorphisms in PE could partially elucidate the mechanisms of susceptibility to disease, and contribute to seeking for new diagnosis and treatment methods of PE. PE can lead to severe complications, and even the death of both mother and fetus. Although the complex pathology is not yet clear, some evidence suggested that the occurrence of PE is related to inflammatory factors. We reviewed the current understandings of roles of cytokines in PE, and provided an extensive overview of the role of single nucleotide chain polymorphisms (SNPs) in the genes potentially underlying the pathophysiology of PE.
Collapse
Affiliation(s)
- Chengcheng Guan
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fei Zhao
- Department of gynecology and obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhencui Yang
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Tang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ling Wang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xueli Li
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lixia Zhang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ziwen Deng
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huabin Hou
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingli Wang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yinglei Xu
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ru Zhang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Lin
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ping Tan
- Department of gynecology and obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of gynecology and obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiguo Liu
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
50
|
Sun M, Jiang H, Meng T, Liu P, Chen H. Association Between TLR4 Gene Polymorphisms and Risk of Preeclampsia: Systematic Review and Meta-Analysis. Med Sci Monit 2021; 27:e930438. [PMID: 34334784 PMCID: PMC8343538 DOI: 10.12659/msm.930438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Toll-like receptor 4 (TLR4) plays a pivotal role in the innate immune response and is hyperactivated in preeclampsia (PE). Several researchers have published conflicting evidence for TLR4 rs4986790 and rs4986791 single nucleotide polymorphisms (SNPs) as risk factors for PE. The present meta-analysis was conducted to obtain a more definitive conclusion about the effects of these SNPs on PE susceptibility. MATERIAL AND METHODS To determine the correlation between rs4986790 and rs4986791 polymorphisms in the TLR4 gene and susceptibility to PE, the PubMed, Web of Science, EMBASE, Chinese National Knowledge Infrastructure, and Chinese WANFANG databases were searched for eligible articles. Statistical analysis was performed with STATA software, version 12.0. Pooled odds ratios with corresponding 95% confidence intervals (CIs) were extracted for assessment of correlation strength. RESULTS We identified 5 studies including 578 cases and 631 controls for the rs4986790 SNP and 4 studies including 469 cases and 457 controls for the rs4986791 SNP, mainly from a White population. The pooled analyses showed no statistical relationship between the polymorphisms rs4986790 and rs4986791 and PE susceptibility in 5 genetic models (all P>0.05). Moreover, the allelic and dominant gene models of rs4986790 and the allelic, heterozygous, and dominant gene models of rs4986791 had high heterogeneity. The sensitivity analysis explored potential sources of heterogeneity and confirmed the findings of this meta-analysis. CONCLUSIONS TLR4 rs4986790 and rs4986791 polymorphisms may not be implicated in PE susceptibility, primarily in a White population. More high-quality studies of genetic associations with PE are warranted.
Collapse
Affiliation(s)
- Manni Sun
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Hui Jiang
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Tao Meng
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Peiyan Liu
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| | - Haiying Chen
- Department of Obstetrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China (mainland)
| |
Collapse
|